Investigator’s Handbook A Manual for Participants in Clinical Trials of Investigational Agents Sponsored by the Division of Cancer Treatment National Cancer Institute Cancer Therapy Evaluation Program Division of Cancer Treatment | | National Cancer Institute (v ©) Bethesda, Maryland 20892 “235283085 Pu Table of Contents aE £ TOA WN bo TOVROONCTION! 510 00510510 0500 5 2.505 5 015 5 5.70 2k 51% mom 1 950 900.010 ac 4 oro goin 450 3 0 7 Part A Sponsors, Research Bases, and the Investigator ................... . . . . . | 9 2. The SPONSOT . . oo... 10 2.1 How NCI Funds Research .................... 00 0. 11 2.2 Preclinical Drug Development ...................................... 12 2.3 Collaboration Between Sponsors: NCI and the Pharmaceutical Industry ..................... .......... 12 2.4 Private Support of Trials Sponsored and Funded by NCI ............... 13 2.5 Private Support of Trials Sponsored but Not Funded by NCI ........... 13 2.6 The Investigational New Drug Application ......................... .. 14 2.7 The New Drug Application ...................cou uuu... 15 3. The Research Base and the Investigator ............................ .. 16 3.1 Definition and Purpose of a Research Base for Clinical Trials. .......... 16 3.2 Activities of the Research Base ..................................... 17 Part B The Development of a Clinical Trial .................. ...... .... .. .. .. ... 21 4. Phase I Trials of Cytotoxic Drugs ........................... IEEE Fhe 22 4.1 Scientific Policies of CTEP ................ 0. i. 2 4.2 Who is Eligible to Study Phase I Drugs ............................. 24 4.3 Which Organizations Can Conduct Phase I Studies. ................... 25 4.4 Who is Eligible to Administer Phase I Drugs ...................... ... 25 4.5 How to Obtain Information About Phase I Drugs. .................... 25 5. Phase II Trials ............. 26 5.1 Scientific Policies of CTEP ................ i. 26 5.2 Who is Eligible to Study Phase II Drugs. ............................ 30 5.3 Who is Eligible to Administer Phase II Drugs ........................ 31 5.4 Restriction on Participation in Phase II Studies ................... .... 31 5.5 How to Obtain Information About Phase II Drugs .................... 31 5.6 The Letter of Intent. .......... 32 6. Phase III Trials. ............ 34 6.1 Scientific Policies of CTEP ................. iii. 34 6.2 Who is Eligible to Conduct Phase III Trials ....................... ... 35 6.3 Coordination of Planning with CTEP Staff ..................... ..... 35 Part C The Planning and Execution of a Clinical Trial ....................... . . .. 37 7. The Drafting of a Protocol ....................................... . 38 7.0 Title Page o.oo 38 7.2 The Essential Elements of a Protocol ................................ 39 7.3 Informed Consent ............ 42 7.4 Model Protocols . ......... 42 8. Protocol Review and Approval at NCI. .............................. 43 8.1 The Protocol and Information Office. ............................... 43 8.2 How to Submit a Protocol .............. 43 83 IRB Approval ...... 44 8.4 Protocol Review ........... 44 8.5 Protocol Approval .......... 46 Table of Contents (continued) Part D 8.6 AMENAMENLS . «© ott ttt tee ee 46 8.7 Reactivation of Studies ..........iut iii 47 B.8 Study SAMS «vvvvnrivs smsms sims AIBA IRIE ERI BITS Hom an ww ww nn 47 9. Ordering Investigational Drugs from NCI ............................ 48 9.1 How to Place Drug Orders... 48 9.2 Particular Points tO NOE .. o.oo ee 49 9.3 Routing of Drug Requests ...: sins nsurrmsnssmimammrnommrmen san snes 49 0.4 Affiliates and Drug Orders... :csivissistnvsmspsmarminme manne nom es 50 9.5 Requests for the Non-Protocol Use of Investigational Drugs ............ 5 0.0 THC OFURIS. ov vr cnvcrmumn vmsmesmoin sasma sme Ridhimamash gos ems es 51 9.7 Requests for Non-Clinical Use of Investigational Drugs ................ 51 10. Responsibility for Reporting of Results to CTEP ...................... 52 10.1 IOEOAUCHION. © «cvvvvmvcmr mama vm rmr emis bs BREE EHEB ES BREESE men 52 10.2 Reporting Requirements of Phase I Trials ............................ 53 10.3 Reporting Requirements of Phase II and III Studies ................... 54 10.4 Retention of Records ....... coun 55 10.3 Reporting T0 IRDBS w.oviviusnmrmimnimsmmemaamomeas enews oman ss sms bu 55 11. Adverse Drug Reactions ...................c..oiiirriiinunnnnnnnnn. 56 11.] Phase 1 Studies. cv mvcm sms nme sos is imi a cREMEE $Rsms s MID sw simn vn 57 11.2 Phase land 111 Studies ..........ci vi iviririvnsesrvrnnrvnmnsnsnnss 57 11.3 Where tO RePOIt. . oot e tte eee 58 11.4 Adverse Drug Reactions with Non-Investigational Drugs. ............... 58 11.5 Adverse Drug Reactions and Routine Reporting of Toxic Events ......... 58 11.6 Actions Taken by CTEP on Reports of Adverse Drug Reactions ......... 58 The Organization of a Clinical Trial ..................................... 61 12. The Investigator and Protocol Chairman: Roles and Responsibility .............. 62 12.1 The INVESHIZAtOT . . oot i tee e ee ee eee eee 62 12:2 The Protocol. CRAITNAN: oss ss mem ms ms amass sess sm meme mem sins nn oom 64 13. Affiliate Investigators. . ............ 65 13.0 Definition ....oviiiiiii iinet tern rinresasarunananaannsens 65 13.2 Requirements of an Affiliate Investigator. ............................ 66 13.3 Responsibilities of Affiliate Investigators .................. nt. 66 13.4 Responsibilities of Research Bases for Affiliates ....................... 67 14. Who May Administer Investigational Drugs .......................... 68 14.1 Restrictions to Physicians Registered as Investigators with CTEP ........ 68 14.2 Administration of Investigatioal Drugs by Non-Registered Physicians... .. 69 15. Accountability and Storage of Investigational Drugs ................... 70 15.1 Procedures for Drug Accountability and Storage ...................... 70 152 Drug RetUINS .....oivvi iii iiiiir inna sansasnneaanasssannnans 71 15.3 Verification of Compliance ...........coviiiiiviniuraenrrerreesenns 71 15.4 Handling of Antineoplastic Agents ...............oooiiiii een. 71 16. Monitoring and Quality Assurance ................... 72 101 INLTONUCHION «sn anno as Bon 53 TEBE HED ANE LES Ewa wa swan #0 da 72 16.2 Protocol Compliance ........ cuir iineearnririarasenanas 22 16.3 DAA ACCUTACY + «vv vr vn sms nn sme sme sens isinensonivonnessnss vanes 73 16.4 Procedural RequUIr€mMents . . .......ouuiuit teas 73 16.5 Components of the Quality Assurance Program Implemented by CTEP ........oivtivnimivrsnivmmnrninccnianimsni ns 74 16.6 Informed Consent and the Monitoring Program. ...................... 76 16.7 Dealing with Problems Identified at Site Visits ........................ 77 Part E Non-Research Use of Investigational Drugs ............................... 79 17: OUOUP TC i ininvini nth imi ime is i oimaibt me rms mr dmn mins ohn nn me has 80 17.1 Definition ...... oi 80 17.2 Placement of a Drug into Group C ................ iii... 80 17.3 Current Group C Drugs ..........couiiii 80 17.4 Use of a Group C Drug .........oo iii 80 17.5 Requesting a Group /C DIU. oc cuv vis iotmsimims sme ns is eosmbanans ne 81 17.6 Responsibilities When Using Group C Drugs ......................... 81 18. Special Exceptions .............. 82 18.1 Definition .... o.oo. 82 18.2 Criteria for Approval of a Special Exception Request .................. 82 18.3 Requesting a Special Exception Drug ...................... 0.0... 83 18.4 Responsibilties of Physicians Administering Special Exception Drugs ............o i 83 APPENdices. . . ..... 85 I. Policy Statement - Relationships Between the Pharamaceutical Industry and Therapeutic Trials in the Clinical Cooperative Groups Supported by the National Cancer Institute. ....................c.vuuurnon... 89 II. Policy Statement on the Conduct of Phase I Trials in Children. ....... 95 I. ‘CTEP Organizational CHAIL. ; cs wuivsnsins stint anim ins ins@edni®s 101 —List of Key Addresses and Telephone Numbers. .................. 102 IV. FDA Statement of Investigator (FDA-1573)........................ 105 V. New Drug Studies Group Application Guidelines. .................. 109 VI. Letter of Intent Submission Form................................ 113 VII. Informed Consent Checklists. .................................... 117 Vill. Protocol Submission CHecKiSIS. ..ocusniamsmiis mr sninsatnsmnsnsm 121 LX: CTD ClOSSATY csv 25 sma 05.05.4655 5 8mm bw em are wesw wns wm wos v tom 0 9 04 000 125 X. Clinical Drug Request (NIH-986)................................. 131 XI. Revised Guidelines for Reporting Adverse Drug Reactions. ........... 135 —Drug Experience Report (FDA-1639). ............................ 141 XII. NCI Investigational Drug Accountability Record Form. ............. 145 XIII. Recommendations for Handling Cytotoxic Agents, National Study Commission on Cytotoxic Exposure. ............................. 149 XIV. NCI Procedure for Management of Investigational Drugs Acquired for Compassionate Treatment of Individual Patients................ 157 1. Introduction Over the past few years the Division of Cancer Treatment (DCT), National Cancer Institute (NCI), has developed policies concerning various aspects of clinical drug development. They are intended to ensure patient safety and to provide the National Cancer Program with the most effective drug development program possible. In this handbook, we explain the policies and procedures of the DCT with respect to the clinical use of its investigational drugs. Some DCT policies are a direct result of the regulatory requirements of the Food and Drug Administration (FDA) and the Department of Health and Human Services (DHHS). Others are not directly mandated by law or regulation but are the result of a consensus among DCT staff, the DCT Board of Scientific Counselors and leaders in the community of clinical investigators. We have also described in this volume the specific procedures that the NCI has in place to implement its policies. Oncologists, nurses, pharmacists, research administrators, and data managers should find the information presented here of assistance in practical matters connected with protocol writing and submissions, reporting requirements, drug account- ability, and a host of other subjects. Specific policies and procedures are constantly in evolution; through them NCI aims to provide a flexible and responsive system within the con- straints imposed by regulation and the size and scope of the program. With respect to drug development, the NCI acts both as a funding agency and as a sponsor (in the sense used by FDA) of clinical research. The DCT has broad responsibility for this effort. Within the DCT the Cancer Therapy Evaluation Program (CTEP) designs and implements the develop- ment plans for new agents. Aside from the purely scientific and medical issues involved in such planning, CTEP utilizes two particular adminis- trative tools: (a) it files Investigational New Drug Applications (INDs) with the FDA permitting DCT to act as a drug sponsor; (b) it is responsible for the contracts and cooperative agree- ments under which most clinical testing takes place. In this handbook we shall refer to DCT as the drug sponsor and the ‘‘proprietor’” of the drug development program. We shall refer to CTEP wherever the efforts of CTEP staff specifically come into play. We shall reserve the use of NCI for more general contexts including overall support of clinical trials. We welcome the reader’s comments on the content of this volume and how future editions may be made more useful. Daniel F. Hoth Robert E. Wittes Part A Sponsors, Research When investigators perform DCT in the conduct of Bases, and clinical trials two Orgsnize: Sinica! Iria ve A ci : tions are crucial—the spon- iscuss the vital role of the the Investigator sor of the trial and the research base in support of research base. The next two the investigator. As is the sections discuss the pur- case with all types of poses and features of each. research, clinical trials In particular we describe cannot take place without the role of the DCT as a a substantial institutional sponsor of investigational commitment. drug trials; as a sponsor DCT is responsible for the overall direction of the process of drug develop- ment, as well as its prac- tical implementation. Of course, DCT is not the only sponsor of trials involving new anticancer agents; the activity of private pharmaceutical firms has increased drama- tically in recent years. We outline here the basis of the relationship among DCT, private firms, and investigators supported by 2. The Sponsor 10 The development of new anticancer agents is a long and complex process. Successes have been significant. The fact that many aggressive neoplasms are now curable with chemotherapy is the best possible evidence that agents with selectivity against cancer can be identified and used effectively. On the other hand, oncologists are well aware that for many common tumors sys- temic treatment is unsatisfactory. The motivation to develop better therapy is therefore as powerful as ever. With the increased understanding of the malig- nant process, due to recent and future advances in molecular biology and biochemical pharmacology, there is every reason to expect that drug development will proceed along increasingly rational lines in the future. The process of drug development is often divided into preclinical and clinical components. Although this division is operationally useful, it should be recognized that a continual interplay and crossfeeding exists between the preclinical and clinial arenas. Evidence of synergy or of the effectiveness of combined modality approaches in experimental models, for example, has provided the major motivation for a very large number of clinical trials. The converse is also true; clinical observations have from time to time given rise to new lines of basic investigation. Historically, the most important effec- tor in the discovery and development of new anticancer agents has been the NCI. The prominence of NCI’s role in cancer drug development has no parallel elsewhere in developmental pharmacology. The justification for such intensive involvement of a government agency in research and development is clear: significant improvement of cancer treatment is in the public interest and in the past there has been insufficient involvement on the part of the pharmaceutical industry. This is in contrast to the impressive role of the private section in the development of many other classes of drugs, such as antibiotics, anti-inflammatory agents, and endo- crine agents. Current trends suggest an increasing interest by industry in anticancer drugs. Even so, NCI re- mains the largest sponsor of research with antineoplastic agents; currently well over 100 compounds are in various stages of clinical testing, and a far greater number are in preclinical development. As part of this massive effort the NCI funds a clinical trials network which includes cooperative groups, new drug development contractors, and other investigators at cancer centers and uni- versity hospitals. More than 5000 investigators from about 500 insti- tutions participate in this effort. In the United States, clinical research with experimental drugs is closely regulated. The ultimate authority for assuring the safety of the public in matters relating to drugs and medical devices rests with the Food and Drug Administration. FDA regulations, which are specific implementations of the Food, Drug and Cosmetic Act, define the terms under which clinical work with experimental agents may proceed. Since these regulations have the force of law, they must be heeded by all those involved in clinical trials with investigational agents, including NCI, pharmaceutical firms, and inves- tigators. An organization or individual who assumes legal responsibilities for = 2.1 How NCI Funds Research supervising or overseeing clinical trials with investigational agents is termed a sponsor. In the United States the DCT and private pharmaceutical firms sponsor such research in cancer. The designation obviously implies a sub- stantial commitment of resources. In addition, the Public Health Service Act mandates a number of safeguards for the rights and welfare of individ- uals who are involved as subjects of the research. Regulations of HHS, administered by the Office for Pro- tection from Research Risks (OPRR) at the National Institutes of Health (NIH), specify the requirements to ensure adequate protections for human subjects. Clinical investigators and institutions taking part in the clinical trials network are responsible for meeting the requirements of the HHS regulations. As sponsor of an investigational drug, DCT, and specifically CTEP, is re- sponsible for seeing that clinical trials proceed safely and rationally from the initial dose-finding studies through to a definitive evaluation of the role of the new drug in the treatment of one or more specific cancer(s). Fulfillment of this goal obviously requires the active participation of CTEP staff throughout the entire process. A full discussion of the means by which the NCI funds research is beyond the scope of this handbook. Whether support by grant, contract, or cooperative agreement, however, the process of peer review is central. Government officials can only provide monies to investigators in the context of mechanisms involving peer review; this process requires formal applica- tion by the investigator and (usually) multiple levels of evaluation. Once an application is approved the NCI cannot provide more funding than is stipulated by the judgment of peer review and the National Cancer Advisory Board. Additional awards can, of course, be made after review and formal approval of a supplemen- tal application. The provision of drugs for clinical studies is a separate issue and does not imply that NCI will provide funding for these same studies. = 2.2 Preclinical Drug Development = 2.3 Collaboration Between Sponsors: DCT and the Pharmaceutical Industry 12 The DCT could not effectively accomplish its overall aims in drug development without a very extensive preclinical effort. The Developmental Therapeutics Program (DTP), and the Biological Response Modifiers Program (BRMP), within the DCT are both heavily committed to the discovery and development of new antineoplastic agents. Readers are referred to a summary of the objec- tives, methods, and status of DCT’s preclinical drug development program in Driscoll, JS: The Preclinical New Drug Research Program of the National Cancer Institute, Cancer Treatment Reports, 68: 63-76, 1984. At present many anticancer drugs in clinical development by CTEP are also being developed by a pharma- ceutical firm. Although the involve- ment of the pharmaceutical industry in research on antineoplastics is not new, the maturing of medical oncology as a specialty and the advent of successful drug treatment of many cancers has stimulated industry’s interest in cancer treatment to an unprecedented degree. Collaboration between DCT and the pharmaceutical industry may occur at any step along the drug development process. Private firms often submit compounds to DCT for testing and joint development. Compounds may be submitted for antitumor screening, for preclinical toxicology, or for clinical testing. Conversely, if a compound is discovered by DCT, the involvement of a private firm is sought as early in development as possible, since DCT does not take drugs to market. The early involve- ment of a pharmaceutical firm permits substantial cost sharing between public and private sectors, and can hasten by several years the availability of effective drugs for all cancer patients. Development plans for a new agent, therefore, are often a collaborative effort between DCT and a private firm. These plans are heavily influ- enced by the findings and opinions of the clinical investigators working with the agent. In this joint effort, DCT and the private sector share the com- mon goal of defining the contribution of a new agent to cancer treatment as expeditiously as possible. However, since the “‘bottom lines’ of DCT and a pharmaceutical firm are clearly different, the DCT may be more will- ing than a private firm to sponsor certain kinds of exploratory trials that will not lead directly to FDA approval of new or extended indications. The three-way relationship among clinical investigators, the DCT, and private industry involves complex problems in coordination, in estab- lishing priorities, and in the allocation of limited resources. To facilitate the necessary interactions, CTEP has developed a policy on the nature of the relationship between the partici- pants. The policy, which formally recognizes the involvement of the private sector in the support of clinical trials, is articulated in a document entitled ‘Policy Statement- Relationships between Pharmaceutical Industry and Therapeutic Trials in the Clinical Cooperative Groups Supported by the National Cancer Institute’ (see Appendix I). = 2.4 Private Support of Trials Sponsored and Funded by NCI = 2.5 Private Support of Trials Sponsored but not Funded by NCI As private support for clinical trials in cancer becomes more widespread, investigators holding grants, contracts, or cooperative agreements from the NCI should carefully consider what are the allowable allocation of re- sources provided by a private sponsor for a trial already having NCI sup- port. Investigators must make certain that Federal funds are not used to cover those costs of research that are also supported by private resources. Grants management personnel at the NIH and auditors from the DHHS are required to scrutinize such arrangements closely and may take steps to recover Federal funds that have been used inappropriately. In the specific case of the clinical cooperative groups the Terms of Award of NCI’s agreements with the groups have been modified to permit them to accept industrial support, provided that industry funds are used for the support of additional costs generated as a direct result of the interest of a pharmaceutical firm in a particular clinical trial. Such costs might include additional laboratory tests or special requiremeats for data collection. In the case of trials funded under Phase I and Phase II/III contracts, the provision of resources for tasks not supported by Federal funds may or may not be appropriate; all such agreements should be submitted to the contracting officer for prior approval. Private support of an unfunded trial sponsored under an IND held by DCT may be appropriate under cer- tain circumstances. However, there should be a written agreement be- tween the protocol chairman and the private firm; this agreement should also be sent to the Chief, Investiga- tional Drug Branch (IDB), CTEP. In general, CTEP will favor the provision of data from trials of this kind to a private firm. These arrangements may not be exclusive, (i.e. may not serve to prohibit the supply of data to another party) however, unless CTEP has pre- viously agreed with the firm that exclusivity is appropriate. In any case the obligations of the investigator to DCT as the sponsor, as detailed throughout this handbook, remain unchanged. 13 = 2.6 The Investigational New Drug Application 14 Any organization seeking to sponsor clinical trials with experimental drugs must first submit an IND to the FDA#* The IND is the legal mechanism under which experimental drug research is performed in the United States. No experimental drugs may be administered to patients for research in the U.S. without an IND. * All sponsors have obligations specified in regulations of the FDA. The DCT, as a component of an agency in the DHHS, is just as accountable as a pharmaceutical firm for meeting FDA’s IND regulations. *The use of the term sponsor is generally reserved for organizations assuming broad responsibilities for the development of a drug. It is also possible, but unusual, for an individual investigator to hold an IND. + An IND must be filed to perform clinical studies under the following conditions: 1. When an investigational agent is manu- factured in one state or country and trans- ported to another state or country for clinical use. 2. When the bulk material (or components of the agent) is manufactured in one state or country and transported to another state or country for further processing, formulation, or for final fill. Although situations arise in which an agent is manufactured and tested within a state, tech- nically, if any component of that clinical product (from the diluent to the vials and labels) is obtained from another state or country, the FDA could require an IND to be filed and all the requirements to be adhered to. The initial IND submission by the sponsor to FDA is a lengthy docu- ment that sets forth the experimental rationale for human testing, including results of animal toxicology studies, manufacturing data, purity and stability information, and an initial plan of clinical investigation. After a sponsor has submitted an IND, the FDA has 30 days to com- plete its review. If FDA has any objections, it may place a hold on the initiation of any or all clinical trials with the agent. In highly unusual circumstances, the sponsor may request a waiver of the 30-day waiting period. Please note that these are matters between the sponsor and the FDA. No investigator may initiate patient treatment on a protocol using DCT drugs until receiving written notice of approval from CTEP. The IND is the official record at the FDA of the sponsor’s clinical research with the drug. Under FDA regulation, CTEP must maintain the IND as an accurate, timely repository of all infor- mation concerning clinical use of the drug, including all protocols, adverse reactions, and an annual report of the results of all clinical trials, plus any new relevant preclinical (particularly toxicologic) data. Obviously this means that there can be no use of the experimental drug without the knowl- edge and prior approval of the SpONSOT. » 2.7 The New Drug Application After clinical trials have shown that the new drug is safe and effective, there is reason to make the drug generally available to patients and physicians. The formal process in the U.S. by which this occurs is the approval by FDA of a New Drug Application (NDA) submitted by a private sponsor; as noted previously the NCI does not submit NDAs since it does not market products. The applicant seeks FDA's approval for one or more specific indication(s). Review and approval of an NDA is based on the demonstration of safety and efficacy assessed from detailed reports of the clinical trials, parti- cularly randomized controlled studies. The contribution of a new drug in the treatment of a disease is demonstrated unambiguously if the drug is the only variable between the treatments. The specific endpoints which consti- tute satisfactory evidence of efficacy (e.g., response rate, quality of life, survival) are a matter of continuing discussion. On the advice of its Oncologic Drugs Advisory Committee (a panel of outside experts in clinical oncology), the FDA has tended in re- cent years not to regard tumor shrink- age per se as important evidence for efficacy in the absence of a positive measurable effect upon survival or quality of life. The subjective nature of many assessments of quality of life has made it an infrequently used basis for an NDA. Clearly the least controversial and most stringent measure of efficacy is a survival bene- fit for the population treated with the new agent. Although FDA regulations do not strictly require that a demon- stration of increased survival be by randomized controlled trials, the FDA's guidelines make clear that this study design is certainly the preferred one whenever feasible. The approval of the NDA is a critical milestone not only for the pharma- ceutical firm but also for the clinical investigator, the practicing oncologist, the NCI, and the general public. An affirmative decision by the FDA permits the pharmaceutical firm to market and promote the drug for the approved indication(s). Once a drug is marketed, no Federal regulation pre- vents any licensed physician from prescribing it for any indication he/she deems appropriate. For the practicing oncologist, NDA approval means that the drug is readily available for routine treatment of patients. The practitioner no longer has to devise protocols without re- search intent, simply for the purpose of obtaining the drug for patient care. No longer must he/she use the cum- bersome procedures for obtaining compassionate INDs from the FDA to treat individual patients. For the clinical investigator, NDA approval means that it may be more difficult to recruit patients for further clinical trials with the drug, since use of the drug is no longer restricted to patients on research protocols. For the NCI, NDA approval marks a step forward in the development of effective cancer therapies. Although DCT’s role as a sponsor of clinical trials focusing on the approved agent usually decreases dramatically at that point, the NCI continues to sponsor further research with commercially available drugs through its general support of clinical trials. For the general public NDA approval means that a new effective agent is now available on the widest possible basis. It is admittedly also true that a drug that was formerly available without cost for research purposes is no longer free to the patient after NDA approval. For financial reasons the DCT has found it necessary to discontinue the distribution of vir- tually all marketed drugs, except for certain clinical trials of particularly high priority. 15 3. The Research Base and the Investigator = 3.1 Definition and Purpose of a Research Base for Clinical Trials A research base is an entity which assumes a broad range of responsi- bilities and functions for the support of clinical trials conducted under its name. Examples of research bases in- clude cancer centers and cooperative groups. The research base supports the investigator in developing, organizing, implementing, and analyzing clinical trials. It assumes responsibility for the quality of the research, both in concept and execution, and has an important role in assuring patient safety. An effective research base enhances the investigator’s research in several specific ways. It provides assistance in developing protocols and obtaining approval by sponsoring agencies. It often offers centralized data manage- ment and statistical consultation. An effective research base should also provide the opportunity for internal peer review and quality assurance. Obviously the research base enhances its own scientific credibility by assum- ing responsibility for the quality of the scientific ideas and the care with which they are tested. These activities may also be an economical way of supporting multiple clinical inves- tigations simultaneously. In short, a research base provides an institutional source of support and assistance for the activities of protocol chairmen and investigators. Clinical cooperative groups are examples of research bases whose functions are described in this section. Many of the same considerations about organiza- tion for support of clinical research apply to any cancer center or single institution. a 3.2 Activities of the Research Base The research base may assist the clinical investigator in many activities and may assume primary responsi- bility in several. 3.2.1 Protocol Development 3.2.1.1 Scientific Review - Many research bases have procedures for review of the science of a clinical trial, either at the concept stage or at the time a protocol is written. This review is distinct from the task of the Institutional Review Board (IRB), which may or may not view as part of its charge a critical scientific review. Ideally, a scientific review assists the investigator in focusing his/her ideas and perhaps in identifying other useful scientific resources within the research base. This process should facilitate research and assist in the testing of new ideas. Careful review may be particularly important for Phase III trials, because of the very substantial commitment of time, patients, and resources involved. 3.2.1.2 Biostatistical Consultation - The design of a Phase II or III trial should be based on sound statistical principles. Issues such as sample size, stopping rules, endpoints, and the feasibility of relating endpoints to objectives are pivotal to a successful trial. Statistical expertise should be provided by a research base. 3.2.2 Protocol Administration - Since most protocols require multiple levels of approval, and since policies of the various sponsoring agencies may differ and change with time, a research base can provide valuable assistance to the investigator in obtaining these ap- provals. Establishment of a centralized mechanism for submitting and track- ing a protocol through the necessary approvals, including the IRB, saves individual investigators a very large expenditure of time and effort that is much better directed elsewhere. A research base with multiple CTEP- sponsored protocols could efficiently assume the responsibility for com- municating status changes, amend- ments, results reports, publications and other pertinent protocol administration information to the CTEP. 17 18 3.2.3 Establishment of an Affiliate Program - The participation of affiliate investigators often contributes to the success of cancer clinical trials. The research base must be as con- cerned about the quality of research performed by its affiliate investigators as that of its own staff. CTEP has established a set of guide- lines to assist research bases in devel- oping a policy toward affiliate investi- gators (see Section 13). Each research base participating in investigational drug trials sponsored by DCT should develop its own affiliate policy in accord with these guidelines. This policy should be submitted to the Head, Quality Assurance and Compli- ance Section (QACS), RAB, CTEP. The most important components of these guidelines are that the research base should, (a) define qualifications necessary for affiliate investigators and (b) periodically review their perfor- mance. Such reviews of performance should include site visits by investi- gators from the research base. 3.2.4 Drug Accountability and Storage - Although FDA regulation places the responsibility for maintain- ing accountability for the use of investigational drugs with the inves- tigator, an institution may assume these responsibilities for the inves- tigators on its staff and assure itself of compliance with Federal require- ments (see Section 15). 3.2.5 Reporting of Results to CTEP - The results of trials involving IND drugs must be reported to the spon- sor. The cooperative groups, as research bases, inform CTEP directly of their results. In all other cases, such reporting is the responsibility of the protocol chairman (see Section 10). 3.2.6 Data Management and Statistics - Since most cancer clinical trials involve professional staff other than the protocol chairman, adequate collection of clinical data is a complex task which must be integrated into the medical practices of the institution. Furthermore, data collection is best done as data are generated; this practice promotes protocol compliance and permits the protocol chairman to monitor the study’s progress. For these reasons, data management organized and supported at the department or institution level is usually more efficient and reliable than that which is left to the individual investigator. Centralized data management is obviously not required of institutions performing NClI-supported trials but its advantages seem clear. In the experience of CTEP’s site visit monitoring program (see Section 16) the quality of execution of clinical trials is better in institutions that provide central support for data management. 3.2.7 Quality Assurance - This term, as discussed in detail in Section 16, applies to three features of the conduct of clinical trials. a Data Accuracy - Is the data in the research record an accurate reflection of the primary medical record? » Protocol Compliance - To what extent was the protocol followed? Are the reported results based on the treatment set forth in the protocol? If not, was there good reason for departing from the protocol and are these reasons fully documented in the research records and any resultant publications? = Procedural Requirements - Were all necessary approvals obtained? Was the informed consent process adequate? A quality assurance program permits the research base to satisfy itself that each participating investigator is fulfilling his responsibilities. It also provides the research base with data about the quality of execution of its clinical research, and it provides the investigator an opportunity to learn from an external evaluation of his performance. In its most constructive form, this process constitutes a peer review of performance and improves the quality of clinical research. The implementation of these activities takes many forms. Section 16 describes the existing procedures of the cooperative groups and CTEP for quality assurance. Although CTEP does not require single institutions to establish internal quality assurance programs, the inclusion of this subject in this section is intended to draw attention to the potential value of this activity for any research base. CTEP staff will assist any research base which desires assistance and advice in developing these programs. 19 Part B The Development of a Clinical Trial The following three sections explain CTEP policies for each phase of clinical investigation with experimental drugs. For each phase we outline our scientific objectives as a drug sponsor. We also describe which physicians are eligible to study and administer investigational drugs. These two aspects of drug use—study and administration—are for- mally quite separate issues. In general, eligibility to study NCI investigational agents is restricted to institutions and physicians approved by peer review; these individuals may in- clude new drug develop- ment contractors, coopera- tive group members, phy- sicians affiliated with approved cancer centers, and recipients of investi- gator—initiated clinical research project grants (ROI, POI). The applica- tion of this general rule to each phase of drug devel- opment is explained in detail. Except in certain explicitly defined circum- stances outlined in the following sections, the administration of experi- mental agents is restricted to these investigators. We also describe how investigators may obtain information about individual experimental drugs and CTEP plans for their development. 4. Phase 1 Trials of Cytotoxic Drugs = 4.1 Scientific Policies of CTEP 22 4.1.1 Planning of Phase I Trials - CTEP prospectively plans the Phase 1 development of each drug. Selection of schedules for clinical trial is based on experimental data (see 4.1.4 for details). Usually each schedule is examined in not more than two studies. From the results of the Phase I and clinical pharmacology studies, CTEP and its collaborating investi- gators decide which schedules are to be taken into Phase II. If the need exists, a second schedule may later be examined comparatively in selected tumors to better define the therapeutic index. 4.1.2 Objectives - Phase I trials determine a safe dose for Phase II trials and define acute effects on normal tissues. In addition, these trials examine the drug’s pharma- cology and may reveal evidence of antitumor activity. Therapeutic intent is always present in Phase I trials; indeed, anticancer drugs are not tested in patients unless preclinical activity studies have already demonstrated evidence of significant activity in laboratory models. Animal toxicology studies carried out prior to Phase I trials provide the investigator with (a) estimates of a starting dose for clinical trials and (b) prediction of the likely effects of the drug on normal tissues. These data provide the investigator with clues which help focus clinical observation of the patient. The dose is increased gradually by some defined procedure until a level is found which produces limiting but tolerable toxicity and/or clear signs of therapeutic activity. Phase I trials define acute effects that occur with a relatively high frequency on normal tissues. Continued careful observation during Phase II and III trials is essential to identify less frequent acute toxic effects, as well as cumulative and chronic toxicities. 4.1.3 Patient Selection - Patients eligible for Phase I must have con- firmed malignant disease which is not amenable to conventional forms of therapy or for which there is no standard treatment. Patients should have normal organ function, in order that the investigator may reliably distinguish drug effects from disease effects. When there is impairment of a major organ, drug treatment may produce increased toxicity because of decreased clearance or additive injury to the organ. Since most cancer drugs will ultimately be used in some patients having impairment of major organ function (particularly cardiac, hepatic, and renal) it is reasonable to explore their use in such patients through Phase I trials explicitly designed to determine safe doses and pharmacology in these settings. CTEP will consider sponsoring such trials selectively after the initial trials in patients with normal organ function. 4.1.4 Schedule Selection - The number of separate schedules studied in Phase I is determined by several factors, including evidence of schedule dependency in experimental in vivo systems, pharmacokinetics, mechanism of action if known, and existing clinical data with similar compounds suggesting superiority of a particular schedule. Drugs which are highly schedule- dependent in preclinical models are usually brought into Phase I on the putatively optimal schedule. Since the correlation between schedule dependency in preclinical models and in the clinic is not firmly established, however, some drugs may be candi- dates for a broader array of schedules. DCT is prospectively evaluating the ability of experimental models to predict the schedule dependency of efficacy, toxicity, and pharmaco- kinetics. For drugs showing no particular schedule dependency in models, two extremes of schedules (e.g., single bolus dose per course and 5-day continuous infusion) are generally examined. 4.1.5 Starting Dose - The starting dose of a Phase I trial, as derived from preclinical toxicology, is 1/10th of the MELDj( (mouse equivalent of the LDjo) in mg/m 2, unless that dose is toxic in any species tested; in the latter case the trial begins at a lower dose that has been shown to produce no more than minimal and reversible effects in the most sensitive species tested. 4.1.6 Dose Escalation - Doses are escalated according to a scheme in which the initial increments are large and decrease rapidly as biologic effects become evident. Most com- monly a modified Fibonacci plan is employed. The exact schema may be affected by the steepness of the dose- toxicity curve in animal models. In all cases the goal is to arrive at the recommended Phase II dose with the fewest number of escalations consis- tent with patient safety; this procedure minimizes the number of patients receiving biologically inactive doses. The DCT is actively evaluating alter- nate methods of dose escalation, based on the use of blood level data (see below). At each dose level a minimum of three patients not previously treated with the new drug should be entered. Escalation to the next level should not occur until the safety of the current level has been established. Usually this means that at least three patients each will have been observed for the entire course interval (e.g., 3-5 wks). 4.1.7 Pharmacokinetics - The role of pharmacokinetics in Phase I is now receiving increasing emphasis, with specific focus on the possible use of such data to guide dose escalation. Human data on the pharmaco- kinetics of a parent compound and active metabolites, taken together with similar preclinical data in vivo, will be extensively analyzed over the next several years of Phase I trials to identify new approaches to dose escalations. This work is being coordinated by the Blood Level Working Group, composed of investigators in several programs of the DCT. Investigators developing Phase I trials should consider pharmacokinetic determinations an integral part of a Phase I study. Since this may be limited by the availability of suitable methodologies, investi- gators should check with the IDB drug monitor before writing a protocol. 23 = 4.2 Who is Eligible to Study Phase I Drugs 24 4.2.1 Contractors - CTEP’s primary resources for Phase I trials are institu- tions awarded contracts for these studies. Principal investigators are selected through competitive peer review in response to periodic solici- tations from DCT* These contracts are usually awarded for 3 to 5 years. DCT has two distinct contract programs for Phase I trials: = Phase I/pharmacokinetics of cytotoxic agents in adults. = Phase I trials of biologic response modifiers in adults. 4.2.2 Ad Hoc Phase I Investigators - Under unusual circumstances, other qualified investigators may be selected by CTEP to perform a Phase I study of a particular drug. Such studies are unfunded (see Section 2.1). Investiga- tors may be selected because of unique expertise or research experience relevant to the drug, the availability of certain patient populations, or labora- tory facilities to perform special studies. In all cases such investigators must have demonstrated competence to conduct a Phase I study with anticancer drugs. Ad hoc Phase | investigators must fulfill all CTEP requirements for trials conduct, as defined in this section, and for reporting of data as described in Section 10. *When NCI seeks offerors for a contract, it issues a Request for Proposal (RFP). Notices of the availability of RFP are published in the “NIH Guide to Grants and Contracts)” which is widely distributed to universities in the U.S. They are also announced in the ‘Commerce Business Daily,” which announces all U.S. Government contract solicitations. An investigator wishing to perform an ad hoc Phase I trial should send a letter to the Chief, IDB, CTEP, outlining the proposed study in concept and identifying the specific capabilities unique to the proposal. Please note that CTEP is not solicit- ing proposals under this heading and will be extremely sparing in approving such applications. Approval is made on a study-by-study basis and does not qualify an investigator for general eligibility to test DCT drugs in Phase I as is the case with the Phase I contractors. = 4.3 Which Organizations Can Conduct Phase I Studies = 4.4 Who is Eligible to Administer Phase I Drugs = 4.5 How to Obtain Information About Phase 1 Drugs Phase 1 trials will be conducted by single institutions. Multicenter trials will not be approved, with the excep- ion of pediatric Phase I studies (see Appendix II, Policy Statement on the Conduct of Phase I Trials in Children). All Phase I drugs will be administered only at the institutions listed on the approved protocol under the super- vision of the protocol chairman. These drugs should not be sent to referring physicians, except with written permission of the CTEP. 4.5.1 Clinical Brochure - This docu- ment contains all relevant information about the drug, including animal screening, preclinical toxicology, and detailed pharmaceutical data. Also included, if available, is a summary of current knowledge about pharma- cology and mechanism of action. CTEP maintains a clinical brochure for each investigational drug. Bro- chures may be obtained from the Head, Drug Regulatory Affairs Section (DRAS), Regulatory Affairs Branch (RAB), CTEP. 4.5.2 CTEP Letter - This newsletter contains announcements of the approval of new drugs for clinical trials by the Decision Network CommitteeX Circulated every 2-3 months, the “CTEP Letter’ is an important means of communication between CTEP and the community of Phase I investigators. It is distributed to a restricted mailing list, including *The Decision Network Committee advises the Director, DCT regarding which drugs will be developed and in what priority order. It is composed of staff members of the DCT. principal investigators of the new drug development contracts, chairpersons of cooperative group and the new agents committees, and selected others. 4.5.3 IDB Physicians - Each DCT investigational drug is assigned to a IDB staff physician, who coordinates its clinical development. Phase I investigators are advised to discuss a proposal with this physician before writing a formal protocol. IDB strongly encourages investigators to submit a Letter of Intent for Phase I trials (see Section 5.6). Relevant telephone numbers and addresses can be found in Appendix III. 4.5.4 Other Information - Phase | investigators should carefully read Sections 7, 8, 10, 11, 12, 15 and 16, much of which is relevant to issues arising with Phase I drugs. 25 5. Phase II Trials = 5.1 Scientific Policies of CTEP 26 5.1.1 Drug Development Considerations 5.1.1.1 Planning and Coordination of Phase II Trials by CTEP - As a spon- sor DCT must devise and implement a plan for Phase II trials of new drugs. An adequate Phase II plan, while conceptually straightforward, is often difficult to execute. A reasonable plan presupposes answers to the following questions: = What doses and schedules that have emerged from Phase I ought to be carried forward into Phase 11? = What diseases should be targeted for testing? = How does the new drug fit into CTEP’s priority list for various targeted disease studies? (For example, do we regard an analog of cisplatin as constituting a higher priority than a novel structure for testing in ovarian or head and neck cancer?) = How does the new drug fit into the priorities of the clinical investigators that form the core of the NCI-sup- ported clinical trials network? =» How can the CTEP assure that each agent receives an adequate test in each disease that is studied? How many studies should be mounted in each disease category? What kinds of patients are suitable for study entry? What are suitable stopping rules for Phase II trials? The plan of Phase II development is prepared by CTEP staff during late Phase I and is announced in the “CTEP Letter’ 5.1.1.2 Single Agent Phase II Studies - A Phase II study determines (a) whether a drug has antitumor activity and (b) estimates the response rate in a defined patient population. In addition, well designed Phase II trials do not permit the entry of more patients than necessary to ensure detection of a medically significant level of activity. Phase II studies are disease-oriented. The various tumor types are tested in Phase II as distinct clinical entities, since each has differing prognostic factors, eligibility requirements, and patterns of responsiveness to a particular drug. As there may be many unknown or uncontrollable factors contributing to variability in outcome, CTEP attempts to sponsor two Phase II trials in each of the tumor types. The goal of these initial Phase II trials is to determine whether the new drug has activity against particular cancers. These trials, therefore, serve as a screen for further study. For this reason, every effort should be made to avoid false results. Although false positive results are certainly undesir- able, false negative Phase II results are especially misleading, since the discovery of a potentially useful antitumor agent may be significantly delayed or overlooked altogether. CTEP has adopted guidelines con- cerning eligibility requirements based on patient characteristics that appear to have a particular impact on likeli- hood of response. Specifically, for initial Phase II studies we currently seek trials which restrict patient eligibility to the minimum extent of prior therapy consistent with ethical medical practice. Protocols for the initial Phase II trials of a drug whose MTD has been well characterized should restrict patient entry in the following ways: » For diseases which currently lack effective systemic therapy (e.g., carcinomas of the large bowel, kidney, liver, and pancreas, as well as malignant melanoma), trials should be limited to patients with no prior chemotherapy. » For diseases in which chemotherapy may cause objective regression of tumors but with little or no impact on survival, entry of patients with no prior therapy will also be sought, whenever possible (e.g., carcinomas of the head and neck, cervix, esophagus, prostate, bladder, stomach, and non-small cell lung). In certain situations which present legitimate clinical dilemmas (e.g., extensive small cell lung cancer, disseminated indolent lymphomas, and breast cancer), patients who have received prior treatment with no more than one regimen may be entered on study, provided that performance status is excellent. = For diseases which are potentially curable with systemic treatment (e.g., acute leukemias, diffuse non- Hodgkin’s lymphomas, Hodgkin’s disease, testicular cancer, limited small cell lung cancer, and ovarian cancer), patients having the mini- mum extent of prior treatment compatible with current ethical standards of care are required. This policy will have the following desirable consequences: s Patients initially entered into phase II trials will have the best chance of benefit from treatment and should be able to tolerate any toxicities of therapy better than patients with poorer performance status, drug- resistant disease, and possible com- promised major organ function from prior chemotherapy. » Fewer patients are exposed to inactive drugs. s The chance of missing potentially active agents will be minimized. Clearly the population of patients defined in this way is highly selected and the results of these initial trials will not necessarily be representative of the drug’s activity in the general population of patients with the disease in question. Once a new agent shows significant activity in this initial, relatively favorable subset of patients, eligibility criteria in sub- sequent studies will permit entry of patients with less favorable prognostic characteristics, so that such patients may have an opportunity to benefit from an active agent. In this second stage of the new agent’s Phase II evaluation, a more accurate assessment of its activity in the general popula- tion of patients with cancer may be obtained. CTEP anticipates sponsor- ing no more than two such studies for each disease in which the drug shows significant activity. 5.1.1.3 Combining Drugs - As a general rule, two or more drugs should be combined when there is definite evidence of the activity of each alone against a particular cancer. We believe that the most rational approach to the development of a new cancer drug dictates that it should not be combined with another drug(s) 27 28 until it has shown reproducible evi- dence of activity in at least two single-agent trials in a disease. Alter- natively, or in addition, combinations may be proposed when there is a rationale firmly grounded on labora- tory evidence that is relevant to the clinical circumstance. In the past much development of drug combinations occurred intuitively; oncologists combined two, three, or more putatively active agents in uncontrolled studies of antitumor effect and toxicity. To be sure some very real therapeutic advances were achieved by this process, but the lack of a systematic and stepwise approach and the frequent absence of proper control groups has often left the oncology community in the uncertain position of not knowing whether results with a particular regimen represented progress or not. More particularly, the overall impact of a new agent on both efficacy and toxicity may remain unclear without a systematic approach. Finally, the process of NDA approval is impeded when available data do not elucidate the specific contribution of the new agent. Clearly any intelligently designed and flexible drug development program must provide room for both approaches. Well conceived small pilot trials testing new hypotheses will always have an important place in the developmental therapy of cancer. We shall, however, continue to pay close attention to the rationale behind all proposed combinations and shall continue to ask whether certain proposals for therapeutic research might not be better approached by a Phase III design rather than Phase II. Although the activity of a single agent is the most common basis for its inclusion in a combination, CTEP will certainly consider other rationales. There may be substantial laboratory evidence of synergy between two cancer drugs. Such evidence is parti- cularly compelling if it is also based on a knowledge of mechanism of action. Alternatively, a drug inert against cancer might be added because of evidence that it alters the pharmacodynamics or pharmacokine- tics of the cancer drug. For example, the nitroimidazoles are being tested as both radio- and chemosensitzers, even though they themselves have no anti- tumor activity. In such cases, CTEP will carefully scrutinize the rationale and evidence offered in support of a proposal based on these kinds of considerations. When combination studies are sub- mitted to CTEP for review, therefore, it is particularly important to make clear the goals, background, and rationale of the proposal. If experi- mental results in the laboratory are the basis for the study, they should be relevant to the clinical circumstance and cited in adequate detail. If pre- liminary clinical results are the moti- vation, they should be similarly cited; unpublished results should be pro- vided as part of the background or in an attachment to the protocol docu- ment. If the trial proposes a feasibility pilot, the protocol should state clearly what kinds of results the investigators would regard as medically significant and where they would propose to go next if a significant result is obtained. We are not requesting a detailed plan of a follow-up study or detailed speculations about likely outcomes. Rather, we are seeking an understand- ing of how the pilot proposal will fit into a strategy of development of the new therapeutic idea. 5.1.2 Individual Protocol Considerations 5.1.2.1 Single Disease Studies - Each tumor type should be considered for Phase II study separately. With few exceptions, this means that there should be separate protocols for each tumor type. If there is a compelling reason for including several under one protocol, such as uncommon tumors, then there should be separate state- ments for each tumor type regarding (a) eligibility requirements, including extent of prior treatment (b) accept- able sites for measurable disease (c) response criteria, and (d) accrual objectives. 5.1.2.2 Eligibility Requirements a Tumor types - For each proposed tumor type there should be separate statements on eligibility. a Prior therapy - Because it is clear that the extent of prior cytotoxic chemotherapy is an important determinant of probability of response, CTEP is seeking trials which restrict patient eligibility to the minimum extent of prior therapy consistent with ethical medical practice (see Section 5.1.1.2). = Measurability of Disease - To define quantitatively the antitumor activity of a drug, patients in Phase II trial must have measurable disease parameters. In certain diseases common sites of involvement are either not bidimen- sionally measurable or the tech- niques for assessment do not permit quantifiable measurement. Under these circumstances tumor response may be evaluated without quantifi- cation by an investigator; in such cases it is particularly desirable that responses be assessed by more than one observer. Examples include bone metastases, lymphangitic pulmonary disease, and many parenchymal brain lesions. a Performance Status - Under most circumstances entry to initial Phase II studies should be confined to patients who are largely ambulatory (ECOG = 2). Patients should be expected to survive a sufficient period of time so that adequate observations can be made. « Organ Function - Evidence that the function of major organs is normal is required. This includes creatinine < 1.5; cardiac function at least Grade 2, pulmonary function moderately compensated, and no neurologic, gastrointestinal or endocrine impairment that would compromise the safe use of the investigational agent. 5.1.2.3 Accrual and Statistical Con- siderations - The accrual goals of a study should be specified in advance with a maximum number of patients stated explicitly. Justification for the target sample size, in terms of preci- sion of estimation or levels of type I and type II error, should be provided. The accrual rate of eligible patients which can be realistically anticipated should be given. Mechanisms should be in place for early stopping of negative trials. 29 = 5.2 Who is Eligible to Study Phase II Drugs 30 The following categories of physicians are eligible to serve as investigators in Phase II trials. 5.2.1 Cooperative Groups - All regis- tered physicians of the cooperative group including those at full member institutions, in Community Clinical Oncology Programs (CCOP), at cancer control, or at affiliate institutions. N.B. A cooperative group may have policies which place further restric- tions on investigator eligibility. 5.2.2 Cancer Centers - Staff physicians at institutions designated as comprehensive or clinical cancer cen- ters by the NCI. Such physicians may be: » Staff physicians within the Center. = Physician members of CCOPs for which that center is the research base. = Physicians affiliated with cancer centers (see Section 13 for further details on the affiliate policies of CTEP). 5.2.3 Affiliates -Physicians affiliated with a research base may participate as investigators on CTEP’s Phase II and III drug trials provided that: ms The affiliation is formalized. Its terms should be in writing and based on the CTEP policies on affiliates. (see Section 13). = Each investigator is registered with CTEP by having submitted a signed FDA-1573 (see Section 12 and Appendix IV). 5.2.4 New Drug Development Contractors - This category includes those with Phase I or Phase 11/111 contracts. 5.2.5 New Drug Studies Group - As stated previously, eligibility to test DCT drugs is generally restricted to institutions and individuals approved by peer review. We recognize however that there may be other clinical researchers highly qualified to conduct these trials, and whose participation might benefit the NCI drug develop- ment program. CTEP has therefore established the ‘‘New Drug Study Group’ as a separate category of eligibility to conduct Phase II trials. CTEP intends that applications for this category will only be approved sparingly and in circumstances in which there is a direct scientific benefit to the DCT drug development program, usually because the appli- cant has a unique resource of some type. To apply for this designation, an investigator should submit a document which sets forth the information specified in Appendix V, New Drug Studies Group Application Guidelines. Approval of this application by CTEP will qualify a New Drug Studies Group to submit protocols for Phase II and Phase III trials. The obligations of investigators approved under this category are precisely the same as for those at cancer centers. 5.2.6 Multicenter Phase II Trials - CTEP expects that Phase II trials will be performed only at the proposing research base. If a protocol chairman wishes to collaborate with other institutions not formally affiliated with his research base, the protocol should include a description of the procedures by which the collaborating institutions will manage the conduct of the protocol and should list on the protocol face sheet each institution and the name of responsible investigator at each. The protocol should specifically address the issues described in Section 7.2.14 = 5.3 Who is Eligible to Administer Phase 11 Drugs = 5.4 Restriction on Participation in Phase II Studies « 5.5 How to Obtain Information About Phase II Drugs For a particular clinical protocol, physicians who may administer DCT investigational drugs are those registered (see Section 14) with CTEP and: = who are members of any research base or formally designated affiliate which is listed on the face sheet of the protocol; and = any others who are individually named on the face sheet of the protocol. Administration of drugs by any other physicians is explained in Section 14. Please note that CTEP may restrict the testing of any investigational drug to a very limited number of locations. Although most drugs proceed to a Phase II program open to all eligible investigators, some are restricted to single centers or to specific centers until a safe, reliable Phase II dose has been defined and CTEP and the investigator community are confident that the drug is ready for general testing among all investigators. These decisions are announced in the “CTEP Letter?’ 5.5.1 Clinical Brochure - This docu- ment contains all relevant information about the drug, including animal screening, preclinical toxicology, and detailed pharmaceutical data. Also included, if available, is a summary of current knowledge about pharmacology and mechanism of action. The brochure contains a full description of the clinical toxicities that have been seen in clinical trials. CTEP maintains a clinical brochure for each investigational drug it sponsors; they may be obtained from the address listed in Appendix III. 5.5.2 “CTEP Letter’’ - This news- letter, published every 1-3 months, contains announcements of prelimin- ary findings of interest. Each issue features several drugs and outlines the interest of CTEP in seeking further trials. The ““CTEP Letter’ is an important source of information for principal investigators of contracts at cancer centers, chairmen of coopera- tive groups and of the groups’ new agents committees, and selected others. 5.5.3 IDB Physician - Each DCT investigational drug is assigned to an IDB staff physician who is responsible for coordinating its clinical development. When an investigator has important concerns about the design of a contemplated trial, he/she should contact that physician. 31 = 5.6 The Letter of Intent 32 5.6.1 Definition - The Letter of Intent (LOI) is an investigator’s declaration of interest in conducting a Phase II trial with a specific investigational drug in a particular disease. Approval of the LOI by CTEP reserves that “slot” for the investigator’s protocol if it is submitted within a defined timeframe (see Section 5.6.7) and signifies agreement that the investigator shall submit a protocol based on the terms stated in the LOI. 5.6.2 Purpose - CTEP has devised the LOI system to maximize the efficiency and fairness by which experimental drugs are allocated to investigators for study. Proper use of the system ensures both CTEP and investigators of a steady flow of new agents into the clinical trials system. It enables CTEP to plan the development of several agents simultaneously. For the investigator, the LOI system also promotes much saving of time and effort, since its use should spare him/her the writing of a protocol that might not be approved. Protocols submitted subsequent to favorable review of an LOI are much more likely to be approved without request for major modification, since many of the crucial features of a Phase 11 proposal must be specified in the LOI itself. The system also provides the investigator with an opportunity to explore the proposal with CTEP staff at the concept stage. 5.6.3 Ground Rules for the LOI System - LOIs should be submitted for all Phase II trials that include a DCT investigational drug. They should be submitted according to the following schedule: = Drugs Beginning Phase II - In late Phase I, the ““CTEP Letter” will carry an announcement of a dead- line for submission of LOIs for the initial round of Phase II trials. us All Other Phase II Trials - After this deadline has passed, investiga- tors may submit LOIs at any time. Each Phase II protocol should be preceded by an approved LOI. How- ever, CTEP will consider any phase II proposal from an investigator eligible to use investigational drugs. Our experience demonstrates that protocols submitted without a previously approved LOI are more likely to be rejected as unnecessarily duplicative or needing major modification. 5.6.4 Submission of LOIs - In order to adequately review the LOI, CTEP must have the following information: ms Drug(s), doses and schedule of administration = Tumor type = Patient characteristics, including extent of prior therapy and performance status = Projected accrual rate and target sample size. Citing recent perfor- mance in studies on the same patient population will be helpful. =» Approximate date of study initiation. s Other studies in the same patient population which will be open simultaneously with the proposed study. If there are such studies, a priority list or flow diagram will speed CTEP review. All of this information should be provided on the LOI Submission Form (Appendix VI) and submitted to the Protocol and Information Office (P10), CTEP. Cooperative group LOIs must be cosigned by the protocol chairman and either the group chairman or the group’s executive officer. Drug Development Contractor LOIs must be cosigned by the contract principal investigator. We encourage protocol chairmen to submit, where appropriate, a letter accompanying the LOI which explains in greater depth the rationale, where not obvious, or any unique features of the study. Such additional explanation is not usually necessary for single- agent Phase II trials but may assist in the review of more complex proposals involving experimental drugs. 5.6.5 Review of LOI’s - On receipt of an LOI, CTEP sends an acknowledge- ment to the investigator. Letters of Intent are reviewed by the CTEP Protocol Review Committee (PRC) within 30 days of submission. 5.6.6 Criteria for Review of an LOI - At the time of LOI review the PRC has available information on other studies in that drug/disease combina- tion by other investigators, and other studies in the proposed disease by the investigator submitting the LOI. The committee considers the following in its deliberations: » Study design, including dose, schedule, and comparison groups, if relevant. a The characteristics of the patient population to be studied, parti- cularly the extent of prior chemo- therapy and performance status. = The feasibility of the projected accrual, including an assessment of the past performance of the investigator in that tumor type. » Competing studies of the investigator in that disease. a All other protocols and LOIs for that drug/disease combination from other sources. = Any unique features to the proposal. A letter of approval or disapproval together with any comments is sent to the proposer. 5.6.7 After LOI Approval - Following approval of a LOI, the LOI source has 60 days in which to submit a protocol. At 45 days a reminder letter is sent notifying him/her that the LOI will shortly expire. After the 60 day period has expired, CTEP will not be bound by previous approval of an LOL. 5.6.8 Information About the Status of an LOI - Further information about the status of a particular LOI may be obtained by calling the LOI Coordina- tor in the IDB (see Appendix III). 33 6. Phase III Trials = 6.1 Scientific Policies of CTEP 34 If significant activity is observed in any disease during Phase II, further clinical trials usually study a com- parison of the new drug with standard (or other experimental) agent(s). If reasonable standard treatment can be defined for the disease in question, we generally wish to know whether the new agent constitutes a significant contribution to the therapeutic arma- mentarium. A variety of trial designs may be suitable, according to the state of the art of treatment in the particu- lar disease. Probably the most satis- factory one is the controlled trial which compares the new drug to a standard single agent or a standard regimen plus the experimental agent to the standard regimen alone. Whatever design is selected, however, an appropriate control group must exist and relevant endpoints must be used to measure relative effects. Of greatest medical importance, of course, are relative survival and quality of life. Other measures, such as complete remission rate or disease-free survival, may also be of interest. These studies, which attempt to isolate the role of a new agent in the treat- ment of a specific cancer, are of obvious importance to industrial sponsors, since the results are pivotal in applications to register the drug for commercial distribution. They are of no less importance to the oncology community, since such approval makes the drug generally available for patient care. The results of such trials may be of great medical importance as well. If the control group is properly selected and the experimental treat- ment is constructed as imaginatively as possible, such trials may yield valuable information for the care of cancer patients. Every protocol must contain a section that discusses the study design and the plan for evaluation of the data. The major objectives of the study should be stated as hypotheses to be tested and a target sample size should be clearly specified. Justification for the sample size goal, in terms of precision of estimation or levels of type I and type II error, should be provided. In a Phase III study, it is insufficient simply to give the number of patients to be accrued on each arm. The protocol should specify the test to be used to compare the treat- ment groups, and the probabilities of drawing incorrect conclusions when performing this test with the proposed sample size should be given. The magnitude of improvement in out- come which can be reliably detected using the planned sample size should also be specified. The accrual rate of eligible patients per year which can be realistically anticipated should be stated and documented. If evaluation of treatment effect will require use of nonrandomized con- trols, a thorough description of the control group to be employed should be part of the protocol. This descrip- tion should include a detailed discus- sion of comparability issues and analytic techniques. = 6.2 Who is Eligible to Conduct Phase III Trials = 6.3 Coordination of Planning with CTEP Staff Phase III trials may be submitted from any source eligible to submit a Phase II trial (see Section 5.2). Since the sample sizes required for such studies are quite large, however, a multicenter approach is frequently the only feasible one. It is expected, there- fore, that the clinical cooperative groups will be the major research bases for such trials. Proposals for Phase III studies from single institu- tions should be very specific in docu- menting adequate accrual potential. Furthermore, if the proposal includes collaboration with institutions not formally affiliated with the research base, the protocol should include a description of the procedures by which the collaborating institutions will manage the conduct of the protocol (see Section 7.2.14). Large clinical trials involve years of effort and a substantial expenditure of resources. Accordingly, a certain amount of coordination is necessary for the optimal planning of specific studies of this type. Staff members of CTEP are in an excellent position to advise investigators on the existence of other proposed or ongoing studies which are closely related or even identical to ones being planned. In addition CTEP staff can advise inves- tigators contemplating large-scale trials concerning the concordance of the proposed trial with CTEP program goals. The intent here is not to control or direct but rather to offer helpful advice. We therefore invite investigators to discuss ideas for large-scale clinical trials with CTEP staff while these ideas are still in the concept stage. The appropriate contact is either the IDB physician responsible for the drug or the CIB physician responsible for coordinating trials in the specific disease (Appendix III). Optimally, investigators should submit a letter describing the hypothesis to be investigated, the general design of the contemplated trial, plus relevant information on accrual capabilities, etc., to document feasibility. CTEP can then formally review and provide a Program response to these concepts, commenting on study originality and programmatic interest. 35 = = a — Part C The Planning and Execution of a Clinical Trial The following five sections provide a detailed descrip- tion of the responsibilities of the investigator for the implementation of a clini- cal trial, from the drafting of the protocol to comple- tion of the study. They are intended to guide both the protocol chairman and the participating investigator and outline NCI policies on the responsibilities of each in the execution of a clinical trial. 7. The Drafting of a Protocol = 7.1 Title Page 38 A protocol is the detailed written plan of a clinical experiment. This section details the essential features of a protocol. Careful attention to the following material will expedite the review of your protocol by CTEP. The protocol’s face sheet is the primary source of identifying information for the Protocol and Information Office (PIO) of CTEP, for the drug distribution system, for the IND file at FDA, and for the listing of the protocol in Physician Data Query (PDQ) System. Each protocol submitted to CTEP, there- fore, must have a title page or face sheet which contains the following items: = Date of document. = Local protocol number (i.e., institution or group number). = Title of study. = Protocol chairman, including his/ her name, institution/cooperative group, address, and phone number. A trainee may not be protocol chairman (see Section 12.2.2). = Full name of institution/group submitting the study. ms List of each participating institution/group. as For DCT-supplied IND drugs, a listing of each drug by name and NSC number. Cooperative groups may summarize by specifying ‘‘all group members’’ or “restricted to ...”” and list institutions. Protocols from sources other than the cooperative groups should specify each institution or site participating in the study, together with a responsible physician and telephone number at each. = 7.2 The Essential Elements of a Protocol Every protocol must include the following elements: 7.2.1 Objective(s) - The objectives should be stated clearly. They generally should be stated as hypotheses to be tested. The study design should be capable of answering the questions posed by the objectives. The statistical section should clearly state how the data will be analyzed in relation to each of the objectives. 7.2.2 Background and Rationale - Sufficient background information should be included so that the rationale for the study is clear. Any unpublished data relevant to the rationale should be included either in this section or, if extensive, as an appendix to the protocol submission. 7.2.3 Patient Eligibility Criteria - The issues here have been discussed previ- ously (see Sections 4.1.3 and 5.1.2.2). Studies with objective response as an endpoint should include clear state- ments specifying whether tumor sites to be followed for response must be measurable, what criteria must be fulfilled to consider disease measur- able, whether evaluable disease is permitted, and if so, at what sites. 7.2.4 Treatment Plan - Specify proto- col treatment clearly so that it can be followed by all medical personnel. 7.2.5 Pharmaceutical Information - For each drug and dose form, include a pargraph(s) on its preparation, dilu- tion, stability, method of administra- tion, and known clinical toxicities. The supplier of each drug should also be specified. (e.g., NCI, pharmaceuti- cal company, commercially available) 7.2.6 Procedures for Patient Entry on Study - Procedures for patient entry, whether randomized or non-random- ized, should be specified. Required information includes the telephone number of the randomization desk or protocol registration, office, and the patient characteristics and stratifica- tion factors (if any) to be provided at the time of entry. Telephone randomi- zation is preferred; however, if another method is to be used, it should be described. Patients should generally be registered on study prior to beginning treatment. 7.2.7 Criteria for Response Assessment - The criteria for scoring responses should be included. These should be specific for both measurable and evaluable disease. Disease-specific criteria are often required and should clearly indicate acceptable means of measurement, i.e. CAT scans, radionu- clide scans, ultrasound, etc. 7.2.8 Monitoring of Patients - Specify how patients will be followed for assessment of treatment toxicity and therapeutic effect. A table of follow- up parameters which incorporates the schedule as described is particularly useful. Protocol authors should carefully review the clinical brochure for all IND drugs to be sure that they have included all reasonable measures to monitor known toxicities. 7.2.9 Dose Modification for Toxicity - The plan of dose change for toxicity should be stated for each study drug. 39 40 7.2.10 “Off Study’ Criteria - Criteria for terminating protocol treatment and/or removing a patient from treat- ment or from study should be specified. 7.2.11 Statistical Considerations - An adequate statistical section discusses the study design in relation to the objectives of the study and the plan for the evaluation of the data. Specifically: a Total sample size (Phase II studies should specify a maximum number of patients). = Error levels (alpha and beta) in Phase III studies. a Differences to be detected for comparative studies. = Size of the confidence interval to be constructed around the estimated outcome. = Estimated accrual rate and/or study duration, with supporting documentation. = Stopping rules, including procedures for monitoring the progress of the trial to implement early termination. = Expected outcome parameters as appropriate (response rate, time to progression, surival times, etc.). a All studies should specify a maximum number of patients. 7.2.12 Records to be Kept - Specify the document on which each of the following is to be recorded, where it is to be sent, and on what schedule. s On study information, including patient eligibility data and patient history. = Flow sheets, or other forms for interim monitoring. » Specialty forms for pathology, radiation, or surgery when required. a Off-study summary sheet, including a final assessment by the treating physician. 7.2.13 Participation - All protocol treatments and observations will be made by investigator-physicians affiliated with a research base (see Section 3) and registered with CTEP. Under certain defined circumstances, it may be appropriate for interim treatments to be administered by certain physicians not registered with CTEP (other than trainees, who are assumed to be under the supervision of a registered investigator). In such cases, the protocol should state: m Precisely what responsibilities these physicians will assume, including response assessment, and toxicity reporting. wu The intervals at which a patient should be evaluated by a physician- investigator at the research base. See Section 14 for further details con- cerning which physicians may actively participate in a clinical trial involving DCT investigational drugs. 7.2.14 Multicenter Trials - If a research base other than a cooperative group wishes to collaborate with other institutions or investigators not affili- ated with its research base, CTEP expects that: s The protocol will be conducted as a single research effort and data from each participating investigator will be included in the analysis of results. s The protocol chairman will be the single liaison with the CTEP Protocol and Information Office. The protocol chairman, or designee will coordinate the development, submission, and approval of the protocol as well as its subsequent amendments, and all other report- ing requirements as described in Sections 8.8 and 10. a The protocol chairman will be responsible for the conduct of the study and the monitoring of its progress; he/she will (a) review all case report forms from each parti- cipating investigator; uncritical acceptance of summary data alone from other institutions is not sufficient, and (b) have ultimate responsibility for all reporting to CTEP. s The protocol will include the fol- lowing information in addition to that specified in Section 7.2: — Title Page: This should include the name of each participating institution and the name of the responsible investigator at each. — Procedures for patient entry: Usually there should be one lo- cation at the institution of the protocol chairman authorized to register patients. If not, then alternative arrangements should be stated, together with an indication of how stopping rules will be implemented. — Records to be kept: The case report forms should be of a common format and in accord with Section 10. — Monitoring: Particular attention should be paid to describing the procedures for monitoring the progress of a multicenter trial including, (1) how adverse drug reactions will be reported, and (2) the frequency by which the required records will be sent to the protocol chairman. — Quality Assurance: State the methods by which the accuracy of the data submitted from the collaborating institutions will be verified. — Drug Ordering: In most cases each participating institution will order DCT supplied investiga- tional drugs directly from CTEP according to the procedures in Section 9. w There will be only one version of the protocol and each participating institution will use that document. It should not be rewritten or modi- fied by any other than the protocol chairman who is solely responsible for formulating protocol amend- ments after CTEP approval. 41 = 7.3 Informed Consent = 7.4 Model Protocols 42 Each informed consent document must be protocol-specific and contain the elements required by Federal regula- tion. These regulations do not specify the language of the document but provide a list of elements which must be addressed in the text of the consent form. A checklist has been included in Appendix VII to assist you. The use of a model informed consent outline by a research base can assure inclusion of the essential elements and permits tailoring of the protocol- specific elements to the needs of individual studies. CTEP will not approve a protocol if its consent form fails to address each of these elements adequately. Protocol authors should be certain that the description of toxicities of each investigational agent is complete and balanced. You should consult the clinical brochure for information about known toxicities. The proliferation of word processors has greatly eased the life of the protocol writer. Since the essential elements of a protocol are standard for a given class of studies, many clinical trials organizations have devised model protocols, the content of whose elements can be altered to suit the needs of a particular study. The use of model documents is an effective way of ensuring a complete protocol. Those who use such models, however, should make certain that inappropriate ‘‘boiler-plate’ text does not get carried over to protocols where it makes little or no sense. Our experience is that disease-oriented background information and statistical sections (sample size estimates and stopping rules) are at greatest risk for the ‘“‘word-processor syndrome?’ 8. Protocol Review and Approval at CTEP = 8.1 The Protocol and Information Office = 8.2 How to Submit a Protocol Within CTEP, the PIO manages the review process and maintains the offi- cial record of all CTEP sponsored protocols, amendments, results, publi- cations, and protocol-related com- munications as well as all protocol submissions for PDQ. More than 7000 protocols are maintained in the PIO. All protocols and related correspon- dence should be addressed directly to the Head, PIO, Landow Building, Room 4C-33, Bethesda, Maryland 20892. The PIO will distribute all mail to the appropriate CTEP physician staff. Please do not direct protocol- related materials to any other CTEP staff member. Doing otherwise will lengthen the time required for resolution. Please accompany the materials relating to a single protocol with a Protocol Submission Checklist (see Appendix VIII) indicating the type of enclosure, i.e., new protocol, revised protocol awaiting approval, study status notice, amendment, and “‘other’” (special communications, study summaries, publications). All telephone calls regarding status of protocol and amendment reviews should be directed to the PIO at 301-496-1367. Each new protocol should be sent directly to the PIO. You must include in the submission: 8.2.1 - The appropriate Protocol Submission Checklist. (There are two separate forms: one for cooperative group trials, one for all other studies. See Appendix VIII for samples; please duplicate as needed.) 8.2.2 - Two copies of a paginated, legible protocol, including a local protocol number. Please be certain the protocol document contains informa- tion about each of the topics listed in Sections 7.1 and 7.2. 8.2.3 - An informed consent document which addresses the elements required by FDA regulation (Appendix VII). If these items are missing or incomplete, the submission will be returned to the protocol source without review. Upon receipt by the PIO, each proto- col is assigned an NCI protocol number. You will receive an acknowl- edgment of your protocol submission with the NCI assigned protocol number. This NCI protocol number must be referenced in all subsequent communications with the CTEP regarding this study. The assignment of this number does not imply approval; only the final approval letter signifies approval and the authoriza- tion to order drugs. 43 = 8.3 IRB Approval = 8.4 Protocol Review 44 Each investigator must meet the requirements of the Federal regula- tions for informed consent (45 CFR 46) and for IRB review and approval. See Sections 12.1.1 and 12.1.2. Except for cooperative group protocols, each protocol must have documentation of IRB approval prior to CTEP approval. Evidence of IRB approval may be submitted to CTEP at any time in the review process. If multiple IRBs are involved, only the approval from the coordinating research base need be submitted for CTEP records; of course, each investi- gator must meet Federal regulations for informed consent (45 CFR 46) and IRB review. Failure of investigators to submit evidence of IRB approval is a major cause of delay in protocol approval. The documentation requirements are satisfied by completion of Form 596, or by specifying: a The study title. a The protocol chairman. The institution’s assurance number, i.e., the assurance number issued by the OPRR, NIH. = Date of IRB review. s The dated signature of an institutional official, usually the IRB chairman. CTEP must review and approve every protocol involving DCT investigational drugs. Each protocol is reviewed for scientific merit, duplication of existing studies, patient safety and adequacy of the informed consent. 8.4.1 Receipt of the Protocol by CTEP - Upon receipt, the protocol is checked for completeness (including legibility, complete pages, presence of required sections, and the informed consent). Each protocol is also ex- amined to verify that the investiga- tor/institution is eligible to conduct a DCT-sponsored study, i.e., has an approved mechanism of sponsorship by NCI (cooperative group, cancer center, contract, new drug study group, RO1/PO01, etc.). Investigator eligibility is explained more fully in Sections 4.2, 4.3, 4.4, 5.2, 5.3, and 6.2. If the protocol is incomplete, or the investigator/institution is not eligible under the proposed category of spon- sorship, then the protocol source is so informed and the study is not reviewed for scientific content. CTEP sends the protocol source an acknowl- edgment of studies that are complete and eligible at submission. 8.4.2 Scheduling the protocol for Review - The PIO schedules the pro- tocol for review by the PRC within 2 to 3 weeks of receipt. Studies are registered, scheduled for review (the second Thursday thereafter), and distributed to reviewers on Friday. Therefore, a protocol arriving Thursday afternoon will be scheduled for review in 14 days; if that same protocol arrives Friday afternoon, it will wait another week for registration and distribution, and require 20 days to reach the review committee. 8.4.3 Review by the CTEP Protocol Review Committee - All protocols are reviewed by the PRC. This committee, composed of the professional staff of the CTEP and chaired by the Asso- ciate Director, CTEP, meets weekly and usually reviews 10 to 20 protocols at each session. Each protocol is assigned a minimum of three reviewers; as many as 4 to 6 may be required for complex multimodality protocols. One of the reviewers is always a biostatistician. The informed consent document is also reviewed by a separate professional reviewer. 8.4.3.1 The Review of the Protocol - The PRC discusses the protocol after hearing the reviews of each assigned reviewer and makes a decision that the science of the study is either: wu Accepted as written. un Accepted with recommendations - The investigator is requested to consider the points raised in the consensus review but is not obli- gated to amend the study. If changes are made prior to activa- tion of the study, the investigator must send CTEP an activation amendment which details any changes in the CTEP-approved document. mn Acceptance deferred pending revisions - The PRC has significant questions about the proposed study. It cannot be accepted unless the investigators satisfactorily address the concerns of the written consen- Sus review. s Disapproved - In the judgment of the PRC the protocol cannot be approved even with major revisions. The PRC disapproves relatively few studies and only does so when it feels that a proposal is unneces- sarily duplicative or fatally flawed in concept, design, or feasibility. 8.4.3.2 The Review of the Informed Consent - The PRC also reviews the informed consent document to be certain that: as The document includes all required elements of informed consent as mandated by Federal regulation (see Appendix VII). = The description of potential benefits and toxicities for IND drug studies is complete and accurate. Any changes made to the consent document resulting from a CTEP review should be made known to the IRB. It is not the intent of the CTEP informed consent review to supplant the review of the IRB. Provided the consent document meets the require- ments of the regulation and law and contains sufficient information to enable an individual to make an in- formed choice, the local IRB approval of the contents of an informed con- sent document generally is to be regarded as determinative. 8.4.3.3 The Consensus Review - After the PRC meeting the primary reviewer generates a ‘‘Consensus Review,’ which states the collective concerns of the PRC. This consensus review, together with a cover letter stating the summary PRC decision, is sent to the protocol source within 40 days of the receipt of a complete protocol. 8.4.3.4. Responding to CTEP Consensus Review - If revisions are required in the protocol or the informed consent, the investigator should send a revised protocol and/or informed consent document to the PIO. The revised documents should be accompanied by a cover letter which details the responses to the points raised in the consensus review. If the consensus reviewer finds the response satisfactory, then the protocol goes forward for final approval. The consensus reviewer may, and often does, choose to send the revised protocol back to the full PRC for further consideration. In any case, if the protocol is still not accepted, then a letter is sent to the investigator detailing any remaining CTEP con- cerns. This process continues until the science and informed consent are each accepted or the study is withdrawn or 45 = 8.5 Protocol Approval = 8.6 Amendments 46 disapproved. Each evaluation of a revised study requires 10-30 days on average. Upon final acceptance of the science and the informed consent, as indi- cated by the concurrence of the consensus reviewer and appropriate CTEP officials, a letter of protocol approval is sent to the protocol source (requires 7-10 days average). Please note that no protocol approvals will be given by telephone. Although CTEP staff may discuss the study with the protocol chairman, he/she should consider nothing official until receiving written notice. After written approval is sent, orders for inves- tigational drugs will be honored. Approved protocols using DCT-spon- sored drugs are submitted to the FDA as part of the IND file. Any change to the approved protocol document must be documented point by point and submitted to the PIO (see Protocol Submission Checklist, Appendix VIII). Please reference the NCI protocol number, date each amendment, and number sequentially for each study. Upon receipt, each amendment is reviewed by a CTEP physician. 8.6.1 Activation Amendment - Any change in the protocol which occurs between CTEP approval and activa- tion by the research base should be submitted to CTEP as an ‘‘Activation Amendment’. Such amendments are often used when the CTEP approval letter conveys recommendations that the investigator wishes to implement. 8.6.2 Scientific and Participant Amendments: » Each amendment which modifies the science, the statistics or the par- ticipants of the approved protocol must be submitted to CTEP for approval prior to implementation. a For those rare studies where an immediate change is imperative for patient safety, the IDB staff physi- cian for that drug may be contacted by telephone and the written amendment must be sent to the PIO within 3 working days. 8.6.3 Administrative Amendments: » Each amendment which clarifies the study by editorial corrections, changes in administrative informa- tion, changes in protocol chairman, or changes in the informed consent must be submitted to the CTEP for recordkeeping purposes and will be acknowledged in writing. Each amendment is recorded in the official PIO protocol file, the drug distribution file, the IND file, PDQ, and by the CTEP Clinical Trials Monitoring Service (CTMS) (where applicable). N.B. Cooperative Group Protocols: Amendments to these studies are handled in accord with procedures arranged directly between CTEP and the Group. The review procedures stated above do not apply to the Cooperative Groups. = 8.7 Reactivation of Studies = 8.8 Study Status 8.7.1. - Protocols which are tem- porarily closed require written CTEP approval prior to reactivating if they are: = temporarily closed for reasons of patient safety = closed for reasons of peer review, site visit, or other NClI-initiated reasons. 8.7.2 - Protocols which are tem- porarily closed to accrual by the research base based on early stopping rules do not require CTEP approval to be reactivated. Changes in study status should be communicated immediately to the CTEP PIO. Changes can be easily reported on the Protocol Submission Checklist (see Appendix VIII). The following list includes the categories of study status recognized by CTEP and PDQ: us Approved - CTEP approves the protocol in writing when the science and informed consent are accept- able, the IRB documentation is on file (not applicable to Cooperative Groups), there are adequate supplies of the requested investigational drug, and when the investigator has a current signed FDA-1573 on file with CTEP. If the approval includes recommen- dations, and they are adopted by the research base, then an ‘‘Acti- vation Amendment’’ should be submitted to indicate any changes to the CTEP approved document. mn Active - After CTEP approval the decision by a Group or Institution to open a study for patient entry. Temporarily Closed - The decision by a Group or Institution to stop patient entry pending study evaluation. Reactivated - Patient entry resumes. Closed - The decision by a Group or Institution to close a study to new patient entries; previously entered patients will continue treatment. Completed - The study is closed and no patients are being treated or followed for data collection. 47 9. Ordering Investigational Drugs From NCI = 9.1 How to Place Drug Orders 48 The DCT will provide investigational drugs for use in CTEP-approved protocols, as well as for Special Exception and Group C guideline requests (See Section 18) to registered investigators with a current FDA-1573 (see Section 12.1). Drugs will not be sent to assistant investigators who are not named on the protocol or who have not filed an FDA-1573. INVESTIGATIONAL DRUGS SUPPLIED BY DCT WILL BE USED ONLY FOR TREATMENT OF PATIENTS ENTERED ONTO CTEP- APPROVED PROTOCOLS. Complete the Clinical Drug Request Form (NIH-986) (see Appendix X) which provides the following information: = Investigator name, and NCI assigned investigator number. » Telephone number of investigator and/or individual preparing form. a NCl-assigned protocol number, cooperative group number, or IND number. as NSC number; current inventory as indicated on Drug Accountability Record of drug name, strength and dose form, and quantity. » The investigator or his/her designee must date and sign the drug request form. No matter who may be delegated to sign this form (e.g. the pharmacist) the investigator is responsible for the disposition of all investigational drug shipped under his/her name. If the requester is at an affiliate institution of a research base, then the request should also be signed by an authorized individual at the research base. Specify your affiliation, such as cooperative group, cancer center, CCOP, etc. Only one drug item may be entered on each line on the form. If there is more than one protocol per drug, use a separate line for each protocol. Indicate the official mailing address to which drug is to be sent. Orders should be submitted to the Drug Management and Authorization Section, IDB. The address is listed in Appendix III. = 9.2 Particular Points to Note = 9.3 Routing of Drug Requests wu The clinical drug request form should be completed in full and typed. If it is not complete, the form will be returned. a All clinical drug requests should use this form including those for Special Exception and Group C use. us Allow three weeks from time of mailing the drug order until receipt. = Do not order more than a 2-month supply at a time. = Avoid ordering excessive quantities. CTEP will reduce the quantity shipped if the order is excessive in relation to protocol requirements, or if the inventory at CTEP is insufficient at that time. ns Telephone requests will be honored only for genuine medical emergen- cies in which a drug is needed immediately for a patient on a protocol. However, we recommend that rush orders be sent via over- night delivery service, which greatly expedites your receipt of drug supplies. All such orders are filled the same day if the order arrives before 12 noon. In general, investigators should submit drug requests directly to the Drug Management and Authorization Section (see Appendix III). There are, however, three special circumstances: 9.3.1 Cooperative Groups - Most cooperative groups require that drug requests be routed through the opera- tions office. Check with your coopera- tive group operations offfice to deter- mine its policies. 9.3.2 CCOPs - Drug orders must be submitted to CTEP through the research base which is conducting the study (i.e., cooperative group or cancer center). 9.3.3 Biologic Agents - Drug orders for biologic agents on Phase I trials sponsored by the BRMP should be sent to the Biologics Distribution Desk, Biologic Resources Branch, BRMP, NCI-FCRF, Building 426, Frederick, Maryland 21701. 49 = 9.4 Affiliates and Drug Orders 50 CTEP strongly prefers to ship investi- gational drugs to the most distal site in order to simplify as much as pos- sible the procedures of drug account- ability. Therefore, investigators at satellites or affiliates of a research base should obtain investigational drugs by submitting a request to CTEP. As noted above in most cases this order should be routed through the research base for verification and authorization. CTEP policy does not permit the transfer of drugs between institutions. CTEP intends that IND drugs be distributed directly to investigators. If under exceptional circumstances emergency transfer seems justified, explicit approval by CTEP is required. In addition, investigators who are involved in late Phase II or Phase III drug studies but who only see a small number (five or fewer) of patients each year may receive drug directly from a protocol chairman or from a member principal investigator in the Cooperative Group. There should be no secondary distribution (i.e., provision of drug from one physician to another) of drugs received for Phase I studies, Group C or Special Exceptions. Finally, CTEP may approve a special distribution arrangement for certain unusual circumstances. = 9.5 Requests for Non- Protocol Use of Investigational Drugs = 9.6 THC Orders = 9.7 Requests for Non- Clinical Use of Investigational Drugs Requests for use of DCT investiga- tional drugs under Group C or Special Exception categories must satisfy certain requirements. These considerations are detailed in Section 18 of this Handbook. Please remember that these requests originate from participating pharma- cies rather than investigators, and must be accompanied by a fully completed Schedule I and a DEA Form 222. DCT anticipates cessation of its THC distribution program in late 1986, as soon as a commercial sponsor is prepared to assume the responsibility. Requests for use of these drugs for laboratory experiments should be directed to the Pharmaceutical Resources Branch, NCI (See Appendix III for address). 10. Responsibility for Reporting of Results to CTEP = 10.1 Introduction 52 The timely and accurate reporting of data from investigational drug trials to the sponsor is an important responsi- bility of investigators testing IND drugs. The receipt of these reports in a timely fashion is not an arbitrary requirement. The information con- tained in them is the material which informs CTEP of the progress of the development of the drug and suggests promising new directions. In addition, the material is required by CTEP to meet its obligations under FDA regu- lations to (a) monitor the study and (b) submit an annual report of current findings to that agency. Failure to comply with these reporting require- ments is a serious breach of the agreement that each investigator makes in signing the FDA-1573 and may result in suspension or termina- tion of investigator privileges. For all trials, the reporting of two types of data is important: individual patient data and study summaries. Each is briefly discussed in the following two paragraphs. Following that, specific reporting requirements for Phase I trials are detailed in Section 10.2, and for Phase II and III trials in Section 10.3. nu Case Report Forms - Information about patients is recorded on case report forms which incorporate all patient data stipulated in the protocol. The case report document should not be the same as the patient’s primary medical record. The former ultimately serves as the formal and fixed data base on which the study is reported. A well-designed case report form will assist the investi- gator and assistants in collecting and recording all data called for in the protocol. The patient’s chart, although the primary medical record, is generally not organized for purposes of research and does not reliably contain the judgments of the treating physician on the effects of the protocol treatment. For these reasons a separate research record (i.e., case report form) should be maintained on each protocol patient. Case report forms are best maintained concur- rently with the medical record. A case record should include not only the actual data, but the re- sponsible physician’s assessment of the treatment effect (e.g., response category) and judgment as to whether any medical events in the patient’s course were treatment- induced (i.e., drug toxicity). Unfortunately toxicities are probably often underreported, particularly if the effect is well-described. Study Summary Forms - The study summary is a tabulation and analysis of the collated individual patient data. It includes not only objective tabulations of data, but the assessment of the protocol chairman concerning each case, with particular attention to eligibility, evaluability, and interpretation of the observations. In addition to data recording and reporting, investigators and protocol chairmen have several other respon- sibilities for the reporting of per- tinent protocol information to CTEP. Procedures for reporting study data vary according to the type of study and category of sponsorship. They are outlined below for the three phases of clinical drug development. = 10.2 Reporting Requirements of Phase 1 Trials 10.2.1 Individual Patient Data - For each patient on trial, data are record- ed on a CTEP case report form. This form is specific for Phase I/11/phar- macokinetic trials. All information specified in the protocol should be recorded on this form. It must be maintained prospectively. These forms are submitted biweekly to the CTMS of CTEP. The biweekly submission should include case report forms on patients actively on study and all new patients entered since the last update. At the end of each course, the investigator should indicate which medical events in the patient’s course were in his judgment drug induced. All evaluations regarding toxicity should be reported according to the CTEP toxicity criteria scale. This is a standard format which has been adopted by all CTEP Phase | investigators. These reporting requirements apply to all Phase I trials of drugs newly entering clinical trial, including both adult and pediatric studies. Investi- gators will receive a full report of their study from the CTMS each month. They should review the printout for accuracy and report any discrepancies to CTMS. The CTMS provides CTEP with summary infor- mation on all trials in the data base with each drug. Investigators may, if they wish, receive this summary information for any drug they are studying. The CTMS analyzes the data from Phase I trials for timeliness of submission and completeness and provides monthly reports of this analysis to CTEP investigators. A microcomputer based software package (ACES) is now available from CTEP which emulates the case report form. Data from Phase I trials may be entered directly in a PC; the software contains a facility for direct transmission via telephone to CTMS. 10.2.2 Study Summaries - CTEP has developed a study summary form which contains the protocol chair- man’s tabulation of results and assessment of the results. This document is designed to be main- tained prospectively. It should be submitted to the CTEP PIO every 6 months and at study completion. 10.2.3 Adverse Drug Reactions - Phase I investigators should be acutely aware of the need to maintain close tele- phone contact with the CTEP drug monitor responsible for the study and inform the drug monitor of any signi- ficant new findings. Since the defini- tion of an adverse drug reaction in Phase I is not clearcut, an investigator should regard any severe, life-threaten- ing, or fatal event as potentially reportable immediately by telephone (See Section 11 for full details). 10.2.4. Study Status - Changes in study status must be promptly com- municated in writing to the CTEP PIO (See Section 8.8). Telephone dis- cussions with CTEP Physician staff are not considered formal notice of status changes. 10.2.5 Amendments - All amendments to a Phase I protocol must be sub- mitted and approved by CTEP prior to implementation (see Section 8.6). 10.2.6 Presentations - A progress report of the study is presented by the protocol chairman at meetings of the Phase I Working Group, held at NCI, usually three times per year. Minutes of the meeting are circulated to Phase I investigators and other interested parties. 10.2.7 Publications - Any publication resulting from a DCT-sponsored study should be sent to the Protocol and Information Office, CTEP, identifying the protocol by the NCI protocol number. 53 = 10.3 Reporting Requirements of Phase II and Phase III Studies 54 Reporting requirements for these studies differ from Phase I in the frequency of reporting and in other respects depending on whether the clinical trials organization is a single institution or a cooperative group. The following apply to all Phase II and Phase III trials. 10.3.1 Individual Patient Data - Currently CTEP is exploring the feasibility of using individual case report forms for experimental drug studies performed in cancer centers and other single institutions. These case report forms are being field- tested by the Phase II-III drug devel- opment contractors. If, as we expect, these forms prove feasible and useful, we shall later widen implementation at single center studies. Where these forms are used, each participating investigator is responsible for assuring that the data from patients under his/her direction will be maintained on these forms. He/she also bears final responsibility for their content, whoever else may assist in the recordkeeping. These documents will then be submitted to the protocol chairman or his assistant for further review. These forms may be audited during a site visit (see Section 16). N.B. Cooperative Groups - Each cooperative group has specific record keeping procedures for its investiga- tors. Each group also has its own specific case report form(s), which must be used on all of its trials. The group’s procedures should be followed in all its trials. 10.3.2 Study Summaries - CTEP is implementing a standard Study Summary Form for Phase II trials which will be maintained prospectively and submitted to the CTEP every 6 months and at study completion. For non-cooperative group studies, this task is the responsibility of the protocol chairman. The Study Summary Form should be signed by the protocol chairman, indicating he/she has reviewed it and is in accord with the assessments in the document. It should be sent to CTEP PIO. If these study summaries prove useful, they will be implemented widely among clinical trials organization performing new drug studies under DCT sponsorship. Their use in Phase III studies is also being explored. 10.3.3 Study Status - Changes in study status must be promptly communi- cated in writing to the CTEP PIO (see Section 8.8). Telephone discus- sions with CTEP physician staff are not considered formal notice of status changes. 10.3.4 Adverse Drug Reactions - The importance of reporting adverse drug reactions (ADR) cannot be overstated. Section 11 discusses these requirements in detail. You are reminded that some types of reactions must be telephoned immediately. IDB maintains a 24-hour line at 301-496-7957 to record these messages. You are urged to call if in doubt concerning the need to report any particular reaction. N.B. Cooperative Groups - Most cooperative groups have established procedures for the reporting of ADRs, which are usually reported to an indi- vidual in the group who is in turn responsible for relaying the informa- tion to others in the group and to CTEP. If you are in doubt, contact the operations office of your group. If you are still in doubt or cannot reach that office, contact the IDB at the number listed above. = 10.4 Retention of Records = 10.5 Reporting to IRBs 10.3.5 Amendments - For investiga- tional drug trials, any change in the approved protocol document must be submitted to CTEP for review. Speci- fic procedures for the submission of amendments and a description of the review of amendments of CTEP are outlined in Section 8.6 10.3.6 Presentations - CTEP usually sponsors two meetings annually at which protocol chairmen present the findings of current Phase II trials. These usually focus on results of single-agent Phase II trials. 10.3.7 Publications - Any publication resulting from an investigational drug study sponsored by DCT should be sent to the CTEP Protocol and Information Office, identifying the protocol by the NCI protocol number. FDA regulations require that all case report records must be kept by the investigator for at least 2 years after an NDA has been approved or the IND has been closed. CTEP will notify investigators when these events occur. This requirement is an explicit part of the FDA-1573 (see Appendix IV). Each investigator must report to his/her IRB any problems, serious adverse drug reactions, or proposed changes in the protocol which may affect the status of the investigation and the willingness of patients to par- ticipate in it. The investigator must also give a report to the IRB at inter- vals appropriate to the degree of risk in the study, but no less frequently than once a year, or at study closure. 55 11. Adverse Drug Reactions 56 Because most antineoplastic agents have a very narrow therapeutic index, toxic effects of treatment commonly accompany drug administration. Since cancer patients often exhibit signs and symptoms referable to cancer or its complications, both the definition and identification of a medical event as an adverse reaction to a cancer drug present special problems for the investigator and sponsor. We have developed operational definitions of ADRs which apply to anticancer drug trials; these definitions are described in Sections 11.1 and 11.2 below. The prompt reporting of ADRs to the CTEP is the responsibility of each investigator engaged in clinical research with investigational drugs supplied by the DCT. Investigators are encouraged to submit reports even if there is only a suspicion of a drug effect. Timely and accurate reporting of ADRs is necessary because only the sponsor can collate information from diverse sources and quickly disseminate the information to investigators working with the drug. The centralization of information on real or suspected ADRs makes possible a much more accurate determination of the degree to which a suspected reaction is in fact drug- induced. Finally, regulations require DCT to report to FDA all findings regarding ADRs occurring in trials under its sponsorship. N.B. Investigators should apply the toxicity scales as defined by their research base in meeting the reporting requirements set forth in the following paragraphs. = 11.1 Phase I Studies « 11.2 Phase II and III Studies 11.1.1 Report by phone to IDB within 24 hours: = All life-threatening reactions (Grade 4) which may be due to drug administration = All fatal events A written report should be submitted within 10 working days. 11.1.2 Report the first occurrence of any toxicity of any grade by telephone within 24 hours. An ADR report may be required. For aplasia in leukemia patients: Grade 4 unknown and known reactions not to be reported. Grade 5 unknown and known reactions reported with 10 working days if clearly related to experimental drug (ie., single agent therapy). Otherwise do not report. Adverse reactions in these studies should be handled according to whether the reaction is known or unknown. The list of known toxicities includes those found in the protocol. If in doubt, consider the reaction unknown. 11.2.1 Unknown Reactions: Grades 2 - 3 should be reported in writing to the address listed in Section 11.3 within 10 working days. Grades 4-5 should be reported within 24 hours to the telephone number listed in Section 11.3. A written report should follow within 10 working days. 11.2.2 Known Reactions: Grades 1-3 should not be submitted as an ADR. They should be included in the routine report of the study by the protocol chairman. Grades 4-5 should be submitted as a written report within 10 working days to the address listed below. Grade 4 myelosuppression need not be reported as an ADR. For aplasia in leukemia patients: Grade 4 unknown and known reactions not to be reported. Grade 5 unknown and known reactions reported within 10 working days if clearly related to experimental drug (i.e., single agent therapy). Otherwise do not report. 57 = 11.3 Where to Report = 11.4 Adverse Drug Reactions with Non- Investigational Drugs = 11.5 Adverse Drug Reactions and Routine Reporting of Toxic Effects = 11.6 Actions Taken by CTEP on ADR Reports 58 By Phone: 301-496-7957 Available 24 hours. By Mail: This address is for ADRs only. Do not send other correspondence to this location. Investigational Drug Branch P.O. Box 30012 Bethesda, Maryland 20814 Use NCI designated ADR form (Appendix XI). Cooperative group investigators should check with the Operations Office and/or the Chairmen of the New Agents Committee about group practices for handling these reports. For studies sponsored or funded by NCI involving commercially available drugs, ADRs should be reported promptly in writing using the standard FDA ADR reporting form (FD-1639), if that type of side effect has not previously been reported in the package insert and/or the literature. A clear description of the suspected reaction should be provided along with an assessment as to whether the event is drug- or disease-related. The report should be sent to: Investigational Drug Branch, PO. Box 30012, Bethesda, Maryland 20814 The reporting of ADRs is in addition to and does not supplant the report- ing of toxicities as part of the regular scientific report of the results of the research protocol. Reporting of toxi- city should be in accord with the procedures for reporting of results described in Section 10. CTEP physicians review each sub- mitted report, including the investiga- tor’s assessment. This review may result in a request for further information from the investigator. Each submitted ADR report is classified according to its likely relation to the drug, and to the patient’s underlying disease. Based on this assessment, a decision is made concerning the need for further action. The prime consideration is whether the new findings affect the safety of patients enrolled in ongoing trials. If so, CTEP takes immediate steps to notify the investigator community and the FDA simultaneously. In addition other measures may be taken, including: = Communicating the new informa- tion by: — Sending written notice directly to investigators. — Publishing findings in the “CTEP Letter.” » Altering existing research: — Modification of protocols. — Discontinuation or suspension of one or more trials. = Investigating the reaction: — Initiate special clinical or preclinical studies. = Altering the process of informed consent: — Modification of existing informed consent forms. — Inform patients of new findings. 59 Part D The Organization of a Clinical Trial The following six sections provide a detailed descrip- tion of NCI policies and procedures for the respon- sibilities of individuals and institutions, including the research base. It includes a discussion of affiliates, accountability and storage of investigational drugs, and monitoring and quality assurance. 12. The Investigator and Protocol Chairman: Roles and Responsibilities = 12.1 The Investigator 62 An investigator is any physician who assumes full responsibility for the treatment and evaluation of patients on research protocols, as well as the integrity of the research data. The term distinguishes physicians acting as investigators from those who assume the more limited role of simply administering investigational drugs. The investigator is responsible to see that the protocol is followed, and the data are collected promptly and accurately, even if he delegates the administration of the drug to another physician. Most cancer trials involve collaboration among many investigators, one or more of whom is designated a protocol chairman as distinct from that of the participating investigators. There are about 5000 physicians eligible to receive DCT-sponsored investigational cancer drugs. Most are eligible because they are investigators supported by a peer-reviewed NCI- funded grant, contract, or cooperative agreement. Each investigator agrees to certain essential principles of partici- pation in clinical drug research. These principles are contained in an agree- ment, the FDA-1573, which is defined by FDA regulation (see Appendix IV). 12.1.1 The FDA-1573 In signing the FDA-1573, the investigator assures CTEP that the clinical trial will be conducted according to ethically and scientifically sound principles. More specifically, a signed FDA-1573 commits the investigator to the following obligations or tasks: s The investigator provides on the FDA-1573 a statement of the education and experience which qualify him/her to perform the study. = The investigator assures that a properly constituted IRB will be responsible for the initial and continuing review and approval of the study. Any changes in the research protocol will require IRB approval, and all unanticipated problems involving risks to human subjects must be reported to the IRB. The investigator is required to maintain adequate drug account- ability records (See Section 15). The investigator is required to prepare and maintain adequate and accurate case histories designed to record all observations and other data pertinent to each patient (Section 10). The investigator is required to furnish reports to the CTEP. (See Section 10). In the case of multicenter studies, it is actually the coordinating statistical center and the protocol chairman who are responsible for the generation of these reports. The investigator is responsible for promptly reporting any ADR reasonably regarded as caused by, or probably caused by the drug and if the ADR is alarming, it shall be reported immediately. CTEP devised a detailed policy statement which adapts this necessarily ambiguous language to the setting of cancer clinical trials, so that an investigation may more easily determine when a medical event needs to be reported to CTEP. (See Section 11). s The investigator shall maintain the records of drug accountabililty and case histories for two years following the date a NDA is approved or if an application is not approved, until 2 years after the IND or study is discontinued. Upon the request of a scientifically trained and properly authorized employee of the DHHS (either FDA or NCI), the investigator will make records available for inspection and copying. The investigator certifies that the drug will be administered only to subjects under his/her personal supervision or under the supervision of physicians responsible to him/ her. A list of these investigators should be provided on the FDA-1573. The drug will not be supplied to any other investigator or to any clinic for administration to subjects. (See Section 14, Who May Administer Experimental Drugs). The investigator certifies that he/she will inform subjects or their representatives that drugs are being used for investigational purposes and will obtain the consent of the subjects or their locally authorized representatives. The investigator assures CTEP that the study will not be initiated until the IRB has reviewed and approved the study. 12.1.2 Responsibilities of an Investigator for Human Subjects Protection - The responsibilities of an investigator for IRB review deserve special mention. Each investigator who participates in NCI sponsored clinical research must have the research approved by an IRB. The procedures followed by the inves- tigator and his institution for the protection of human subjects, including IRB composition and function, as well as the basic elements of the informed consent document are specified in regulations of the DHHS. All clinical research sponsored by the DHHS must be in compliance with these regulations (Title 45, Code of Federal Regulations, Part 46). Within the DHHS, the Office for Protection from Research Risks (OPRR) at the NIH administers these regulations with each institution. The OPRR negotiates assurances of compliance with HHS regulations for the protection of human subjects. Under an assurance the IRB is authorized to review and approve research. Each investigator who participates in NCI sponsored research must conduct the research at an institution with an OPRR approved assurance. If this assurance is lacking a single page agreement may be filed with OPRR. Further information about this may be obtained by contacting OPRR at (301)-496-7005. 63 = 12.2 The Protocol 64 Chairman 12.2.1. Responsibilities - The chairman of a clinical trial assumes certain responsibilities in addition to those of the participating investigator. Specifically these include: = Writing the protocol document. = Assuring that necessary approvals are obtained, including those of the IRB, the sponsor (DCT), and any others for the protocol and subsequent amendments. = Monitoring the study during its execution, which includes: — Reviewing each case record to confirm eligibility. — Reviewing each case, record to determine compliance with the protocol. — Reporting of adverse drug reactions. — Determining any necessary changes in the protocol and the informed consent docu- ments and submitting them as protocol amendments to the research base and to CTEP. — Monitoring accrual to the study and stopping the study when the requirements of the study design have been fulfilled. — Reporting study status changes to CTEP (see 8.8). = Analyzing Results - The protocol chairman should make an assess- ment of each case to determine eligibility, evaluability, toxicity, protocol compliance, and out- come. This assessment should be independent of that of the treating investigator. = Reporting Results - The results should be analyzed and tabulated for submission to CTEP. The protocol chairman bears the primary responsibility for this task. In multicenter trials, of course, the statistical center is the protocol chairman’s most impor- tant collaborator in fulfillment of reporting requirements. 12.2.2. Who May Serve as a Protocol Chairman - Since the protocol chairman is responsible for meeting all NCI requirements for IND drug research, as stated in this manual, the protocol chairman must be at the faculty level. Trainees may not serve as protocol chairmen. The only exception is that a trainee may be a co-protocol chairman provided that a faculty member is also co-protocol chairman. In all cases the faculty member is responsible to NCI for meeting all requirements stated in this manual. 13. Affiliate Investigators = 13.1 Definition The participation of physicians who collaborate with major institutions in clinical trials is an important compo- nent of the NCI program. The contri- butions of these participants is recognized by NCI, the clinical cooperative groups, and many cancer centers. In order to assure and main- tain the high quality of clinical research and patient care conducted by the clinical trials organizations, it is important to maintain a strong relationship between these affiliate investigators and the research base. To accomplish this objective, methods of organization are needed that permit all participating investigators to have easy access to accurate and timely information on matters of scientific importance and to conduct treatment research in compliance with Federal regulations. The following guidelines have been provided to assist research bases in formulating specific policies for strengthening the relationship between affiliate investigators consistent with these guidelines. The content of the guidelines applies to affiliates of any research base. We recommend that each research base develop an affiliate policy consistent with these guidelines. An affiliate investigator of a research base is a physician who: = Participates in research clinical trials organized by the research base, and s Has satisfied all criteria for affiliate membership as defined by the research base. 65 = 13.2 Requirements of an Affiliate Investigator = 13.3 Responsibilities of Affiliate Investigators 66 Must have demonstrated competence in the treatment of cancer patients as defined by the research base. Must have the ability to accrue a minimum number of patients as set by the research base. Must have established a close cooperative professional relation- ship with the research base through regular participation in group meetings and/or educa- tional sessions sponsored by the research base. Must have successfully passed a probation period during which time the affiliate investigator has demonstrated: — Ability to enter patients on protocol. — Ability to comply with the protocol. — Ability to adhere to the procedures and standards of the research base and the CTEP. Must have an OPRR assurance for the protection of human subjects (See Section 12.1.2). The affiliate investigator must adhere to the procedures of the research base and CTEP for the conduct of clinical research by: Meeting the recordkeeping policies of the research base. Making certain that each protocol is approved by an authorized IRB prior to involvement of human subjects. = Making certain that each patient signs and is given a copy of the IRB-approved consent form. The consent forms should be main- tained on file by the affiliate investigator. Complying with the policies of CTEP and the FDA concerning the use of investigational drugs which include as a minimum the following: — Filing a signed FDA-1573 with the CTEP. — Observing DCT procedures for the use of investigational drugs, including maintaining NCI Drug Accountability Records (Appen- dix XII). — Agreeing that primary medical records of patients may be au- dited in accordance with the policies of the research base, the CTEP, and the FDA (See Sec- tion 16). = 13.4 Responsibilities of Research Bases for Affiliates 13.4.1 The Cooperative Groups The cooperative groups have the following responsibilities for their affiliate investigators: = Maintaining an accurate and up-to- date list of all member institutions and affiliate investigators. s Informing CTEP of important actions regarding membership status of its member institutions and affiliate investigators. = Reviewing performance of all members in periodic and timely fashion. = Assuring that all members, full and affiliate, are in compliance with CTEP policies and HHS regulations. 13.4.2 The Principal Investigator Responsibilities for Affiliate Investigators in Cooperative Group Protocols The principal investigator in coopera- tive groups has the following respon- sibilities for affiliate investigators: = Assuring the cooperative group that the affiliate member’s per- formance meets the procedures and standards of the research base. = Informing the cooperative group of important changes in affiliate- member relationships. = Agreeing to site-visit the affiliate as deemed necessary by the cooperative group. s Providing the affiliate with accurate and timely information on matters of scientific importance. = Communicating to affiliates in a timely manner all policies (and any changes in policy) on the conduct of clinical research. . 13.4.3 Cancer Centers The cancer centers have the following responsibilities for their physician members and for their affiliate institutions: = Maintaining an accurate and up-to- date list of all members and affiliates. s Informing CTEP of important actions taken regarding membership status of its members and affiliate investigators. » Conducting periodic and timely review of the performance of all members and affiliate investigators. » Agreeing to site-visit the affiliate investigator in accord with CTEP policies. = Assuring that all members and affiliate investigators are in compliance with CTEP policies and FDA regulations. « Communicating to affiliate investigators in a timely manner all policies and changes in policy on the conduct of clinical research. 67 14. Who May Administer Experimental Drugs = 14.1 Restrictions to Physicians Registered as Investigators with CTEP 68 The distribution of investigational drugs is restricted to physicians who are registered as investigators with CTEP. Specifically, the secondary distribution of investigational drugs from registered investigators to unregistered physicians is prohibited except for the clearly defined circumstances outlined below. All patients on clinical trials involving the use of investigational drugs must receive all treatments with these drugs from a registered investigator, except as stipulated explicitly by the protocol that is approved by CTEP. The reason for this policy is clear. Physicians who treat patients as part of a clinical trial must have a com- mitment to the goals of the trial and to the methodologic requirements of clinical research. They must be experienced in the evaluation of therapeutic results and of the toxic manifestations of anticancer therapy. The major issue, therefore, in the conduct of a clinical trial is: Who is making the observations and the decisions? For example, who is deciding on the dose modifications and the laboratory tests to be obtained? Who is interpreting the response data? Who is observing the patient for toxicities of the inves- tigational drugs? A physician per- forming these tasks with investiga- tional drugs must have the training and experience to function effectively as a clincial investigator, and acknowledge his commitment to those principles. All investigators participating in trials sponsored by CTEP must be formally registered with their research base (i.e. cooperative group or cancer center) and with CTEP. Such registration signifies that the physician-investigator meets certain requirements for participation in clinical trials. Investigators register with CTEP by completing a Form FDA-1573. (Appendix IV). Physicians in training may administer investigational drugs under the direct supervision of a registered investiga- tor holding a current FDA-1573. They need not submit a FDA-1573 under their own signature. In such cases the registered investigator assumes com- plete responsibility for the use of these drugs. CTEP recognizes that it is often convenient for a cooperating group member or cancer center physician to ask a local physician to adminster protocol treatment to a patient who may have traveled long distances to the research base for initial con- sultation. We believe that this approach is ultimately detrimental to the clinical research effort unless very careful surveillance is maintained by the investigator. If close monitoring is impossible, it seems much more sensible to require that all treatments be administered by registered investigators. Physicians will have to carefully consider whether a patient being evaluated for study will be able to receive each treatment at the hands of a registered physician. We are confident, however, that the benefits of this policy, in terms of both patient safety and integrity of the research data, far outweigh the disadvantages and are in the long- term best interests of both patients on clinical trials and the drug development program of the DCT. »« 14.2 Administration of Investigational Drugs by Non-registered Physicans For trials involving certain agents which are at advanced stages of development and for which the clinical toxicities are well charac- terized, it may occasionally be appropriate for investigators to seek the assistance of local physicians in the patient’s community to adminis- ter portions of protocol treatment. CTEP will consider this practice to be in compliance with its drug dis- tribution policies provided that all of the following stipulations are met: 14.2.1 - The registered investigator remains responsible to the DCT and the FDA for meeting all investigator obligations listed in this handbook. Remember that you, and not the local physician, are the investigator. You are responsible legally and scientifically for all treatments, whether you administer them yourself or whether they are administered under your direction by an oncology nurse, an oncology fellow, or a referring physician. 14.2.2 - The local physician administers treatment only under explicit instructions from the registered investigator. 14.2.3 - Decisions about dose modification are made only by the registered investigator. 14.2.4 - Observations concerning response status, toxicities, and the decisions on whether to continue or terminate therapy are to be made only by the registered investigator. 14.2.5 - If non-registered physicians are to administer any portion of protocol treatment involving experimental drugs, the protocol document, when submitted to CTEP for review, must state: » Precisely what responsibilities non-registered physicians will assume in drug administration. » The intervals at which a patient will be evaluated by a physician- investigator at the research base. 69 15. Accountability and Storage of Investigational Drugs = 15.1 Procedures for Drug Accountability and Storage 70 The investigator is responsible for the physical storage and recordkeeping of investigational drugs received from CTEP. Specifically, the investigator must: = Maintain a careful record of the disposition of all drug received from CTEP, using the NCI Drug Account- ability Record Form (DARF) (Appendix XII). = Store the drug in a secure location, accessible to only authorized person- nel, preferably in the pharmacy. The intent of the drug accountability procedures described in this section is to assist the investigator in making certain that drugs received from DCT are used only for patients entered onto an approved protocol. The recordkeeping described in this section is required under FDA regulation. Investigators are responsible for the use of investigational drugs shipped in their name. Even if a pharmacist or chemotherapy nurse has the actual task of handling these agents upon receipt, the investigator is the respon- sible individual and has agreed to accept this responsibility by signing the FDA-1573. 15.1.1 - Each drug should be stored separately for each protocol. If a drug is used for more than one protocol, there should be separate physical storage for each protocol. (Remember that CTEP provides drugs on a protocol-by-protocol basis and maintains for each investigator distinct records of drug shipments on each protocol). 15.1.2 - In parallel with the physical storage of drug by protocol, there should be a separate DARF for each drug in the protocol. If there is more than one protocol using a particular drug, there should be a separate DAREF for each protocol. 15.1.3 - Separate accountability forms should be maintained for each different strength or dosage form of a particular drug (e.g., a drug with a 1 mg vial and a 5 mg vial would require a different DARF for the 1 mg vial than for the 5 mg vial). 15.1.4 - The DARF has been designed for use at each location where drugs are stored, e.g., main pharmacy, satellite pharmacy, physician’s office, or other dispensing areas. 15.1.5 - The DARF is also designed to accommodate both dispensing records and other drug transaction documentation (e.g., receipt of drug, internal transfers, returns, broken vials, etc.). A full explanation, as well as a copy of the DARF may be found in Appendix XII. = 15.2 Drug Returns = 15.3 Verification of Compliance = 15.4 Handling of Antineoplastic Agents Many investigators are not aware that drugs should be returned to the sponsor (DCT). Investigators are required to return drugs if: = The study is completed or discontinued ms Drugs are outdated = There is an obvious excess in inventory ms The drug is damaged (i.e., loss of refrigeration) To return drug to DCT: (1) package the drugs securely to prevent breakage, (2) complete the Return Drug List Form, which can be found on the reverse of the Clinical Drug Request Form (NIH-986) (Appendix X), and (3) send to the NCI Clinical Drug Repository at the address indicated on the Return Drug Form. Investigators are reminded that compliance with procedures to ensure proper drug usage will be reviewed during site visits conducted under the monitoring program. Specifically, site visitors will check that the drug accountability system is being maintained, and will spot check the drug accountability records by comparing them with the patients’ medical records to verify that the drugs were administered to a patient entered onto the recorded protocol. There has been considerable concern about the potential risk of chronic exposure to low-level concentrations of antineoplastic agents among health care workers routinely handling these agents. The potential mutagenic activity of antineoplastic agents has been examined in vitro and in vivo. Urinary alkylating and anthracycline agents have shown mutagenic activity in experimental systems, whereas this has not been demonstrated for most of the antimetabolities and vinca alkaloids. Recent reports indicate that antineoplastic agents may be absorbed by workers who are handling them. In addition, some of the compounds are carcinogenic in animals and are suspected of being so in humans, but only in patients receiving the drug at therapeutic levels. However, there is no clear evidence at this time that chronic exposure to low level concentrations of antineoplastic agents has been carcinogenic in health care workers. Nevertheless, it would seem prudent to consider the adoption of certain precautions in the procedures of workers handling these agents. Several professional organizations have reviewed the data on this subject in an attempt to develop guidelines for safe handling. While there are now several published sets of guide- lines, they do not differ significantly. We have reproduced in Appendix XIII the Recommendations for Handling Cytotoxic Agents by the National Study Commission on Cytotoxic Exposure. Please note that these are guidelines and do not have regulatory or legal force. They are included for your consideration and information. 71 16. Monitoring and Quality Assurance = 16.1 Introduction = 16.2 Protocol Compliance 72 Sponsors and research bases must monitor their clinical trials. In assuring the quality of data, monitoring is a key component of any clinical trials program. Quality assurance and monitoring are concerned with the execution of a trial, rather than its conception, and with the quality of the data which support the scientific conclusions. Many individual activities are part of quality assurance, and investigators have recognized some of them as vital to the integrity of clinical trials for years. In particular the quality control of pathology and radio- therapy has been part of the coopera- tive group program for a long time. More recently, investigators have increasingly recognized the impor- tance of verifying the accuracy of other classes of data. We shall now discuss in more detail the items which form the major focus of the DCT-sponsored quality assurance effort. Note that the first two classes of concern (protocol compliance and data accuracy) are really central problems in clinical trials methodology. The fact that they are assessed intensively by the site visit monitoring program should in no way divert attention from their essential importance to the scientific content of clinical trials. 16.2.1 Cooperative Groups - The groups have recognized the importance of assessing extent of protocol compliance for many years. One of the first areas to receive attention was the confirmation of diagnosis. Today virtually all groups have Pathology Committees or Reference Panels for selected studies; central pathology review reduces one important source of variability in trial results. Furthermore, most cooperative groups have quality control in radiotherapy, which consists at least of reviews of port films by group radiotherapists. These reviews are best done prospectively, so that errors can be detected in time to alter subsequent treatment. In the cooperative groups the medical oncology committee or the protocol chairman reviews case report forms to establish whether dose adjustments have followed protocol guidelines, and whether appropriate study tests have been obtained. In most cooperative groups, the protocol chairman also reviews each case to determine eligibility, evalu- ability, and validity of response and toxicity assessment. In some cases, the statistical office accomplishes one or more of these tasks. All of these assessments are performed through review of submitted case report forms. 16.2.2 Cancer Centers - Few cancer centers have organized procedures to assess protocol compliance centrally and systemically (see Section 3, Research Bases). However, the CTEP site visit monitoring program evalu- ates protocol compliance as part of its monitoring site visits. Indeed, this is the major focus of a monitoring site visit to a cancer center. 16.2.3 CTEP Clinical Drug Develop- ment Contractors - Protocol compli- ance is assessed by the CTMS. Phase I and Phase 11/111 contractors submit data to CTMS, which reviews it carefully for extent of compliance with the protocol. Reports of these evaluations are provided monthly to the investigator and to CTEP. = 16.3 Data Accuracy = 16.4 Procedural Requirements The importance of verifying the accuracy of the basic data elements used in the analysis of treatment effects is obvious. Data accuracy is assessed in site visits by comparison of the research record (e.g., flow sheets) with the primary patient record. Response assessment may be evaluated by examination of radio- graphs or scintiscans, where relevant. In many of the site visits performed thus far, CTEP has been concerned about the absence of formalized procedures within many centers for assessing these important issues inter- nally. Many institutions lack central registration mechanisms to enlist patients on trials. Centralized systems of data management are often not available. Some institutions lack clear procedures for certifying the accuracy of research data. Formal procedures for evaluating the accuracy of response assessment, for example by second party review, are commonly lacking. As institutions recognize the importance of these tools for the conduct of clinical trials and bring them “‘on line)’ the quality of data will improve commensurately. As an IND sponsor, the DCT must verify that its investigators adhere to the various procedural requirements of investigational drug trials. The specific procedural activities checked at the time of the site visit are: s Informed Consent - Investigators must be certain that the patient signs the IRB-approved version of the informed consent. Please note that the consent form must be the document specifically written for the protocol. = IRB Approval - Each protocol must be approved by the IRB responsible for the institution where the patient is treated. There should be written verification of this action and of at least annual review. Substantive protocol amendments must also be approved by the IRB. = Drug Accountability - Each investigator must assure that: — All IND drugs supplied by DCT are used only for patients on the specific protocol for which CTEP has approved the drugs. — Logs are maintained which record the disposition of each unit of drug received from CTEP for the protocol. Site visitors may also review the completeness of reporting adverse drug reactions and the quality of recordkeeping with particular reference to the completeness of the source documentation. Each of these areas is reviewed at the time of site visit. In addition, many cooperative groups and cancer centers maintain internal procedures to assure that IRB approvals are obtained prior to patient accession. 73 = 16.5 Components of the Quality Assurance Program Implemented by CTEP 74 16.5.1 Monitoring Monitoring includes following the overall progress of the study to ensure that projected accrual goals are met in a timely fashion and excessive accrual is avoided, that eligibility and evaluability rates do not fall below acceptable standards, and that risks of the study do not outweigh benefits. Poor performance in any of these areas is cause for concern. Because these activities are performed during study execution, they may lead directly to improved conduct of the trial. The cooperative groups are perform- ing these tasks according to systema- tic, formalized procedures. For Phase I studies, the CTMS performs these duties. Cancer center studies are monitored by CTEP physicians through review of semiannual study summary reports. 16.5.2 The Site Visit Auditing Program 16.5.2.1 Purpose of Site Visit Audits = Verification of data accuracy - By comparison of the primary medical record with the case report form maintained by the research base for analysis. = Verification of the presence of an IRB approved consent form signed by the patient. = Verification of IRB approval of each sponsored study. = Verification that procedures for drug accountability, including NCI Drug Accountability Records, are being maintained in accord with DCT procedures. 16.5.2.2 Outline of Audit Procedures = Trials to be audited are those involving DCT investigational drugs. = All site visits to be conducted by teams of peers. s Site visits will be randomly timed. = Site visits will be conducted at an average rate of once every three years (except Phase I trials). 16.5.2.3 Adaptions of Basic Procedures to Specific Needs These basic procedures have been adapted to the several types of clinical trials organizations supported by NCI in the following way: a Cooperative Groups - Each group will perform its own program of site visits, to be conducted by its members. CTEP will attend a reasonable percentage as observers. a Clinical Drug Development Contractors - Site visit audits are made to the Phase I contractors three times each year and to the Phase 11/111 contractors once annually. All are site visited by the CTMS. a Others - This category comprises all others performing DCT IND drug studies, including cancer centers, RO1/POI1 holders (con- ducting clinical IND drug trials as part of grant-related activity) and new drug studies groups. Site visitors will be drawn from a pool formed by investigators at all the institutions to be site visited. Table 1 summarizes the relationship between the content of the site visit audits and the type of clinical trials organization. Table 1 Components of the Quality Assurance Program Protocol Compliance Data Accuracy Procedural Contract CTMS SVAP CTMS Cancer Centers SVAP SVAP SVAP Cooperative Groups Group * Group* Group * * Conducted as part of the site visit auditing SVAP = Site Visit Auditing Program (periodic program of the cooperative groups, and co- site visits for the purposes described site visited by NCI. in the text). + Protocol compliance is verified by each CTMS = Clinical Trials Monitoring Service cooperative group through its own quality (CTEP monitoring organization, assurance program. Most of this is which monitors Phase I and some accomplished through a central review of Phase II trials, and coordinates Site case report forms. However, protocol Visit Auditing Program). compliance may also be checked at the time of site visits to review patient charts, since information pertinent to the eligibility and evaluability of a patient may be discovered which was not included on the case report form. 75 = 16.6 Informed Consent and the Monitoring Program 76 Many have asked about the legality of review of a patient’s primary medical record by outside individuals. The answer is straightforward. No Federal law prohibits external review of a patient’s medical record. The regulations of informed consent do require, however, that the patient be informed of ‘‘the extent to which confidentiality of records will be maintained!” This means that there is no rule against chart review by outsiders, but that the patient must be told what will be done. For this reason, CTEP requires that each informed consent document for investigational drug trials include a statement with the following language “A qualified representative of FDA and NCI may review my medical records?’ Also please note that medical records are protected from inquiries under the Freedom of Information Act (FOIA). Even if the study is per- formed under government sponsor- ship, any records on the premises of the investigator are not subject to FOIA requests. Furthermore, any patient-related records in government files are protected from FOIA requests by the Privacy Act. As an additional measure of safeguarding, CTEP removes patient names from any documents in its possession. = 16.7 Dealing with Problems Identified at Site Visits CTEP and the cooperative groups have a full range of options in dealing with problems identified at the site visit. In a great majority of cases, the measures are intended to be constructive, educational, and corrective rather than punitive. The actions that are taken vary with the individual case. All site visit reports are sent to CTEP. The reports are assessed by CTEP staff on a regular basis. When major problems are identified by the audit team on a cooperative group trial, this information is immediately conveyed to the group chairman and CTEP for further action and investi- gations. After requesting a written clarification, and following review of the case by the cooperative group and/or CTEP, appropriate measures will be applied if the original assess- ment is confirmed. The options for action include: — Letter of Warning — Probationary status — Suspension of patient entry privileges — Immediate repeat site visit — Removal of access to investigational drugs — Notification of FDA if investigational drugs are involved. FDA may conduct its own investigation. The following actions are taken only in instances of fraud or severe misconduct: — Replacement of principal investigator — Termination of grant or contract — Reanalysis or retraction of published results — Formal NIH investigation — Debarrment of investigator from future participation in NIH clinical trials 77 17. Group C « 17.1 Definition = 17.2 Placement of a Drug Into Group C « 17.3 Current Group C Drugs « 17.4 Use of a Group C Drug 80 Investigational drugs which have been given Group C designation by FDA have reproducible efficacy in one or more specific tumor types. Such a drug has altered or is likely to alter the pattern of treatment of the disease and can be safely administered by properly trained physicians without specialized supportive care facilities. If a drug meets the criteria in the preceding paragraph, CTEP may initiate a formal application to the FDA to authorize Group C distribu- tion for the specific indication. Such authorization is not equivalent to formal FDA approval of effectiveness for this indication. A physician should request drugs under Group C if he/she wishes to treat a patient with an indication specifically authorized for the requested drug, as listed in Table 2. All other non-research requests for clinical use are considered a ‘‘Special Exception’ (see Section 18). The usage of a drug for a Group C approved indication is fully described in the GUIDELINE PROTOCOL. This document is written by CTEP and approved by FDA. It describes the indications, dosage, precautions, warning, and known adverse effects of the drug for the specific indica- tion(s). It also contains an FDA- approved informed consent which must be used if there has been no local IRB review. Guideline protocols may be obtained by sending a written request to the Drug Management and Authorization Section, IDB, at the address listed in Appendix IV. Additional information, is available by phone at the number listed in Appendix III. Please note that: a Patients treated under Group C guidelines are not part of a clinical trial at the time of the treatment with the Group C drug. = A drug classified in Group C for a certain indication may still be under active clinical investigation for the same or other indications. a Group C must not be used to obtain drug to conduct clinical trials. = 17.5 Requesting a Group C Drug = 17.6 Responsibilities When Using Group C Drugs = You may request a Group C drug by contacting the Drug Management and Authorization section, IDB, by telephone or in writing. (See Appendix III for number and address.) = You should indicate the diagnosis of the patient. = You must have a current FDA-1573 on file with CTEP (i.e. signed within the last 12 months). » If your request is approved you will receive a supply of drug, and under separate cover, an approval letter, a copy of the Guidelines Protocol, and a copy of the ADR Reporting Form. uw Drug Accountability: You should record the disposition of drug received on the Drug Account- ability Record Form (see Section 15 and Appendix XII). mn Adverse Drug Reactions: Since Group C drugs are still investiga- tional agents, physicians using them are obligated to report adverse drug reactions to CTEP (see Section 11). wu Informed Consent: You must obtain written consent from the patient. The Guidelines Protocol contains the required consent form. ms IRB Review: Since administration of a drug under Group C guide- lines is not done with research intent, there is no CTEP require- ment that IRB approval must be obtained. CTEP has obtained an exemption from this requirment from FDA. You should, however, check with your IRB to determine whether its local policies require approval. In the future, CTEP will consider Group C classification only for those agents whose activity is well enough established that NDA approval in the relatively near future is considered likely. Table 2 *There are special requirements for ordering THC. If you are not familiar with these program, contact the Drug Management and Authorization Section, IDB. Current Group C Drugs Drug Indication Azacytidine Refractory Acute Leukemia Single Agent Treatment AMSA Refractory Acute Leukemia Single Agent Treatment Erwinia Asparaginase Patients Allergic to E. coli Asparaginase Hexamethylmelamine Refractory Ovarian Cancer Single Agent Treatment THC* Chemotherapy-induced nausea and vomiting refractory to standard antiemetics 81 18. Special Exceptions = 18.1 Definition = 18.2 Criteria for Approval Of A Special Exception Request 82 Physicians with patients who are refractory to standard measures, who are not eligible for an ongoing research protocol, and who have a cancer diagnosis for which an inves- tigational drug has demonstrated activity may receive the drug from CTEP as a ‘‘Special Exception” to the policy of administering investi- gational drugs only under a research protocol. If the requested drug is approved for Group C for that indication it should be requested under the Group C Guidelines. (see Section 17.) The Special Exception mechanism is the functional equivalent of a com- passionate IND but differs from it in that the investigator may obtain drugs directly from CTEP, instead of having to obtain an IND from FDA. CTEP provides this mechanism as a service to the oncologic community and to cancer patients. Substantial effort on the part of CTEP profes- sional and support staff is commited to maintaining it. We expect that patients treated under the Special Exception mechanism are not eligible for established research protocols. Drugs available for Special Exception are always in Phase II or Phase 111 trials. Special Exceptions are not granted for Phase I drugs. The purpose of the Special Exception mechanism is to make non-approved agents which have a significant activity against specific malignancies available to cancer patients and investigators. Physician staff members of the IDB review each Special Exception request and make the decision on a patient- by-patient basis, based on the following considerations: » Is there a research protocol for which the patient is eligible? = Have standard therapies been exhausted? » Is there objective evidence that the investigational drug is active in the disease for which the request is being made? A review of the experience with Special Exception Protocols in the past indicated that patients experi- enced considerable toxicity with little significant benefit. As a result, CTEP has attempted to improve the selection criteria for patients treated under Special Exception. Considerable evidence must attest to the activity of the agent for the requested indication. There should be sufficient data available to provide a reasonable expectation that the agent will prolong survival or improve the quality of life in a cohort of similar patients so treated. Reports of low response rates, or responses of brief duration, or anecdotal reports of an occasional response are not sufficient to justify approval. = 18.3 Requesting A Special Exception Drug: = 18.4 Responsibilities of Physicians Administering Special Exception Drugs = Is the drug likely to benefit this patient? Even if the drug has been reported to be active in the disease, the specific circumstances of the patient must be weighed by both his/her physicians and CTEP physicians. Please note that the Special Exception service may not be used as a means to obtain drugs to treat a series of patients on protocol, or to do pilot work for an intended study. CTEP tracks these requests and will take whatever measures are necessary to discontinue such practices by an investigator. (Similar considerations apply to Group C). Drugs distributed under the Special Exception service remain investigational and are subject to FDA regulation and CTEP policy. Requests for Special Exceptions may be made in writing or by telephone to the Special Exception Coordinator, Drug Management and Authorization Section, IDB (See Appendix III). You should indicate the patient’s diagnosis, previous cancer therapy, current clinical status, intended dose and schedule of the requested drug and any proposed concomitant cancer drugs or other therapies. You should explain why the proposed use of the investigational drug is a better selection than a commercially available drug. In addition, the age, sex, and date of diagnosis should be included. (see Appendix XIV). 83 i x Policy Statement: Relationships Between Pharmaceutical Industry and Therapeutic Trials in the Clinical Cooperative Groups Supported by the National Cancer Institute I. Introduction This policy statement pertains to trials involving new drugs or biologicals conducted by the clinical cooperative groups supported by the National Cancer Institute (NCI). It gives general guidelines for the three-way collaboration between clinical investi- gators in the cooperative groups, the pharmaceutical companies, and the NCI. Its contents rest upon the following premises: A. The NCI recognizes the impor- tance of the pharmaceutical indus- try in the clinical development of new anticancer agents. NCI wishes to foster collaboration with indus- try wherever possible. As part of its mission to improve cancer care, NCI shares with industry the im- portant goal of defining the precise contribution of a new drug or biologic in the treatment of cancer. NCI therefore recognizes and supports the need of a private sponsor to focus at the appropri- ate time on clinical trials which lead to a New Drug Application (NDA), since an NDA is the vehi- cle through which new drug therapies become widely available to cancer patients. Thus NCI feels that it is appropriate for the cooperative groups to do clinical trials of interest to, and partially supported by a pharmaceutical firm, provided that the trials have scientific merit and are consistent with the overall goals of the group. The primary consideration for the group in choosing to per- form a trial will be the scientific merit of the proposal and not the availability of additional funds from private sponsors. . Inasmuch as NCI functions as a coordinator of a large volume of clinical research in new drugs and biologics, industry recognizes NCI’s need to be aware of in- dustry’s plans for the clinical development of new agents of mutual interest, particularly if a pharmaceutical firm plans to utilize the resources of the NCI- supported clinical trials mechanism. Industry also recognizes the necessity of preserv- ing the spirit of free and open in- quiry among clinical investigators. . The Clinical Cooperative Groups recognize that their operating pro- cedures remain as defined by the Terms of Award of their cooperative agreements with the NCI. The Groups also recognize that industry may have special needs for data acquisition over and above those of trials not sup- ported by industry. 89 90 II. Planning of Clinical Trials When there is a private sponsor, if the drug to be developed is also of interest to NCI, then the overall plan for its clinical development shall be a collaborative undertaking by the phar- maceutical firm and the NCI; such a plan shall be formulated and/or discussed in joint meeting(s) before the implementation of large-scale clinical testing. In addition to areas of mutual interest, NCI and the pharma- ceutical firm may independently pur- sue clinical studies of particular inter- est to each. Because such studies have implications for commitment of resources by both NCI and the phar- maceutical firm, they shall also be the subject of joint discussion and plan- ning between the NCI and the private sponsor. There should be frequent and full interchange between staff members of the Cancer Therapy Evaluation Program and the private sponsor. Furthermore, whenever possible, the planning of a particular clinical trial, particularly large-scale studies and/or trials of pivotal impor- tance, should be a joint venture in- volving the cooperative group, the pharmaceutical firm, and the NCI. III. Clinical Protocols Protocols involving investigational drugs or biologics in the cooperative groups, regardless of the IND spon- sor, will be activated only after review and approval by the Protocol Review Committee of the Cancer Therapy Evaluation Program. Protocols developed under NCI-IND will be sent to the appropriate private sponsor for scientific review and comment; review by the private sponsor will take place before activation of the study and the comments by the firm will be sent to the investigators along with the CTEP Consensus Review. In addition, large- scale trials will be examined carefully by NCI and the firm for appropriate- ness of resource allocation. IV. INDs Generally the needs of both NCI and the pharmaceutical firm are best served where each holds an IND. We therefore expect that in most cases either NCI or the pharmaceutical firm will file an IND which references an IND or master file held by the other. In certain instances, when agreeable to both the NCI and the pharmaceutical firm, clinical trials within cooperative groups may be conducted under the IND of the pharmaceutical firm. As a general rule, all information in INDs will be fully shared between the National Cancer Institute and the pharmaceutical firm. However, certain information pertaining to manufac- turing processes may be held in confidence by the private sponsor. Nevertheless, all quality assurance data concerning the manufacturing process will be shared. NCI and the pharmaceutical firm will exchange all information submitted to its IND including but not restricted to protocols, adverse drugs reactions, toxicology findings, and other materials. The primary data relating to certain sensitive laboratory studies will, upon request by the pharmaceu- tical firm, be returned to the company by the NCI. NCI files will, however, contain summaries of all such studies. V. Resources Provided to Cooperative Group by Private Sector In principle, the NCI encourages support of clinical research from the pharmaceutical industry. Funds, support personnel to perform additional monitoring, computer resources, etc. may be provided directly to the cooperative group. Funds provided to a cooperative group for additional monitoring beyond the standard practices of the group will be regarded as supplementary funding. As is the case with NCI support, all resources, financial or otherwise, provided to a cooperative group by a pharmaceutical firm should be a matter of record. VI. Data Rights, Confidentiality, and Publications All data derived from clinical trials done by an NCI sponsored group will be made fully available to the NCI. Similarly, data generated in trials directly supported by a pharmaceutical firm will be available to the firm in a manner to be decided upon jointly by the group and the firm. Such data may include case report forms as well as summary data. Data arising from cooperative group trials involving drugs or biologics supplied by a firm but for which the firm has not committed other resources may be made available to the firm by the cooperative group; if there are additional costs to the group for providing such data, the group may be reimbursed by the firm in a manner to be negotiated by the group and the firm. A firm supporting a clinical trial with drugs, biologics, or financial resources committed specifically to that trial shall ordinarily be the only frim to which the group will provide data generated in the trial. The firm may, if it wishes, release the group from this obligation; such a release must be in writing and would permit the group to provide data from the trial to another company. As is the case with all the clinical trials data of a cooperative group, the data generated in trials supported by a pharmaceutical firm are the property of the cooperative group. As stated above, however, these data are to be fully available to the firm and may be used by the firm for its own analyses and in its applications for registration. The cooperative group maintains the full right to present and publish the data at such time and place as its membership sees fit. Manuscripts from pivotal trials or those to which industry has specifically committed financial resources should have advisory review and comment by the private sponsor a meaningful length of time prior to submission for publication. VII. Cooperative Agreement Clinical trials conducted with agents from a pharmaceutical firm irrespec- tive of IND sponsorship or funding arrangements should be included in the progress reports and competitive renewal application of the group. Progress reports and renewal applications should clearly indicate the IND sponsor, if other than NCI, and the resources (funding, personnel, etc.) which have been provided by private SpONSOT. 91 i Policy Statement: The Conduct of Phase 1 Trials in Children In a Phase I trial it is important that each treatment of an individual patient with the new agent be based on full knowledge of the toxicities encountered by other patients at the dosage to be employed. The principal investigator should have access to all clinical data, and has the final responsibility for the conduct of the study. For these reasons, it is NCI policy that Phase I trials should be conducted within a single institution under the direct supervision of a single investigator. Because of the rarity of cancer in children, most pediatric Phase I trials cannot be done efficiently by a single institution. For this reason, the NCI recognizes the appropriateness of conducting multi-institutional Phase I trials in pediatric populations. However, the principles noted in the first paragraph impose special responsibilities on investigators conducting multi-institution Phase I trials. The protocol chairperson must have complete up-to-the-minute knowledge of entire toxicity data set (all patients) in order to most safely and appropriately administer each course of drug. This should be true not only for decisions to escalate doses but also for each retreatment. Obviously, the flow of information between the participating institutions, the protocol chairman and the NCI must be timely, accurate, and accessible. Based on these precepts, the following general policies apply to the conduct of Phase I trials under NCI sponsor- ship in pediatric populations: 1. Selection of Cooperative Group Institutions for Participation in Phase I Trials: Participation should be limited to institutions as follows: ¢ Intensive care and blood bank facilities must be available on site. e Must have at least two full time investigators both willing and judged capable by the New Agents Committee of doing Phase I studies. e Patient entry only at investigator’s institutions; cancer control, CCOP, and satellite institutions may not participate. ® Must have PI approval. e Must have data manager available to collect data as is generated, i.e. daily. 2. Protocol Development and Approval: Drugs to be studied in Phase I trials will be agreed to by the Group and Investigational Drug Branch in the framework of the Phase I Working Group. It is recommended that a Letter of Intent be submitted before developing a full protocol. Protocol authorship, group review and submission to NCI will proceed according to normal group procedures. D3 96 3. Study Chairperson: A protocol chairperson shall be appointed by the Group’s New Agents Committee for each study. Protocol chairpersons should be experienced in the conduct of Phase I trials. Inexpe- rienced investigators interested in Phase I trials may enter the system as participants provided their institutions meet the criteria in (1) above. The Protocol Chairperson is responsible for: — approving each patient entry onto the trial as well as to subsequent levels of dose escalation, — approving each course of treatment for each patient, — deciding when to escalate dose levels, and to what dose, — establishing that the MTD and a Phase II dose have been satisfactorily defined, — terminating the study in a timely manner, — keeping all investigators informed of possible toxicities, — keeping NCI informed of the progress of the trial, — the timely review of data, — the analysis of trial results, — informing NCI of Adverse Drug Reactions. 4. Participating Investigators are Responsible for: — obtaining approval from the protocol chairperson for each course, — assuring that all drugs will be administered at the approved institution, — reporting toxicities and data 5. Data Flow: a) The protocol chairperson must have direct and immediate access to current patient information. Each Group will decide how to triage the flow of data from the investigators to the Group Operations Office and to NCI. The raw patient data should be submitted in the NCI/CTMS data base by posting at biweekly intervals, irrespective of the source of the submission to CTMS. b) Flow Sheets, Criteria for Toxicity and Response Group flow sheets will be transcribed onto standard Phase I case report forms and submitted to CTMS. Criteria for toxicity and response should be the same used by the adult Phase I contractors with any necessary modification for the pediatric population. 6. Adverse Drug Reaction (ADR) Reporting: The responsibilities of reporting ADRs to NCI will rest with the protocol chairperson. Severe life-threatening or fatal toxicities should be reported immediately by telephone to NCI as per the NCI/ADRC reporting guidelines. 7. Site Visit Monitoring The Clinical Trials Monitoring Service (CTMS) will be employed by NCI to audit the data collected in Phase I trials. Periodic site visits will be conducted to compare the patient records with submitted data. This is the standard for adult Phase I studies, and is a reflection of NCI’s obligations as a sponsor of new agents. Because of the number of institutions involved in pediatric Phase I trials a modified site visit schedule will be employed. 8. Drug Distribution Although it is an NCI policy to ship drugs directly to each registered investigator under any specific protocol an exception will be made in this case. The Protocol Chairperson will be solely responsible for drug dispensing and should order directly from NCI. He/she will subsequently re-distribute the drugs to the partici- pating institutions. This increase in authority of the Protocol Chairperson is intended to assist him/her in meeting the increased responsibilities under this policy for Phase I trials. Drug accountability logs should be maintained by each participating investigator. 97 Cancer Therapy Evaluation Program Office of the Director Dr. Wittes - 9 ) Program Management Protocol and and Analysis Office Information Office Ms. Carpenter Ms. Hogan Z ( ] Investigational Drug Regulatory Affairs 1 1 Biometric Research Clinical Investigation Branch Branch Branch Branch Dr. Hoth Dr. Shoemaker Dr. Simon Dr. Friedman Biologics | Evaluation Drug Regulatory Medicine Section Section Affairs Section — Dr. Killen ; Dr. Shoemaker : Dr. Hawkins Developmental Quality Assurance Nutrition and - Chemotherapy and Compliance A Supportive Care Section Section Section Dr. Leyland-Jones Dr. Macfarlane (Vacant) Drug Management _ and Authorization ~ Surgery Section Section Dr. Avis Mr. Fortner _ Pediatric Section Dr. Ungerleider 101 List of Key Addresses and Telephone Numbers 102 FOR ADMINISTRATIVE INQUIRIES Protocols and Amendments: Protocol and Information Office Cancer Therapy Evaluation Program Landow Building, Room 4C-33 Bethesda, Maryland 20892 Telephone: (301)-496-1367 Use this address only for Express Mail: Protocol and Information Office Cancer Therapy Evaluation Program Landow Building, Room 4C-33 7910 Woodmont Avenue Bethesda, Maryland 20814 Letters of Intent: LOI Coordinator Protocol and Information Office Cancer Therapy Evaluation Program Landow Building, Room 4C-33 Bethesda, Maryland 20892 Telephone (301)-496-7957 Adverse Drug Reactions: Investigational Drug Branch P.O. Box 30012 Bethesda, Maryland 20814 Telephone (301)-496-7957 Clinical Brochures: Regulatory Affairs Branch, CTEP Landow Building, Room 4C-17 Bethesda, Maryland 20892 Telephone: (301)-496-7912 Clinical Drug Orders, Group C Drugs, and Special Exceptions: Drug Management and Authorization Section Investigational Drug Branch, CTEP Landow Building, Room 1304 Bethesda, Maryland 20892 Telephone: (301)496-5725 Requests for Non-Clinical Use of Investigational Drugs: Chief, Pharmaceutical Resources Branch Developmental Therapeutics Program Landow Building, Room 5C-25 Bethesda, Maryland 20892 Telephone: (301)-496-8774 Quality Assurance, Site Visits, Monitoring, and Informed Consent Issues: Quality Assurance and Compliance Section Regulatory Affairs Branch, CTEP Landow Building, Room 4C-19 Bethesda, Maryland 20892 Telephone: (301)-496-8798 Regulatory Questions: Chief, Regulatory Affairs Branch Cancer Therapy Evaluation Program Landow Building, Room 4C-19 Bethesda, Maryland 20892 Telephone: (301)-496-7912 FOR SCIENTIFIC INQUIRIES: Biologics Related Questions: Senior Investigator Biologics Evaluation Section Investigational Drug Branch, CTEP Landow Building, Room 4C-19 Bethesda, Maryland 20892 Telephone: (301)-496-8798 Chemotherapy Drug Related Questions: Senior Investigator Developmental Chemotherapy Section Investigational Drug Branch, CTEP Landow Building, Room 4C-09 Bethesda, Maryland 20892 Telephone: (301)-496-1196 Disease Related Questions: Disease Coordinator Clinical Investigations Branch Landow Building, Room 4A-04A Bethesda, Maryland 20892 Telephone (301)-496-6056 DEPARTMENT OF HEALTH AND HUMAN SERVICES PUBLIC HEALTH SERVICE Form approved OMB No. 0910-0013 FOOD AND DRUG ADMINISTRATION STATEMENT OF INVESTIGATOR Note: No drug may be shipped or study initiated unless a completed statement has been received (21 CFR 312.1(a)(12)). TO: SUPPLIER OF DRUG (Name, address, and Zip Code) NAME OF INVESTIGATOR (Print or Type) DATE NAME OF DRUG Dear Sir: The undersigned, submits this statement as required by section 505(i) of the Federal Food, Drug, and Cosmetic Act and §312.1 of Title 21 of the Code of Federal Regulations as a condition for receiving and conducting clinical investigations with a new drug limited by Federal (or United States) law to investigational use. 1. THE FOLLOWING IS ASTATEMENT OF MY EDUCATION AND EXPERIENCE: a. COLLEGES, UNIVERSITIES, AND MEDICAL OR OTHER PROFESSIONAL SCHOOLS ATTENDED, WITH DATES OF ATTENDANCE, DEGREES, AND DATES DEGREES WERE AWARDED b. POSTGRADUATE MEDICAL OR OTHER PROFESSIONAL TRAINING. GIVE DATES, NAMES OF INSTITUTIONS, AND NATURE OF TRAINING. c. TEACHING OR RESEARCH EXPERIENCE. GIVE DATES, INSTITUTIONS, AND BRIEF DESCRIPTION OF EXPERIENCE. d. EXPERIENCE IN MEDICAL PRACTICE OR OTHER PROFESSIONAL EXPERIENCE, GIVE DATES, INSTITUTIONAL AFFILIATIONS, NATURE OF PRACTICE, OR OTHER PROFESSIONAL EXPERIENCE. e. REPRESENTATIVE LIST OF PERTINENT MEDICAL OR OTHER SCIENTIFIC PUBLICATIONS. GIVE TITLES OF ARTICLES, NAME OF PUBLICATIONS AND VOLUME, PAGE NUMBER, AND DATE. IF THIS INFORMATION HAS PREVIOUSLY BEEN SUBMITTED TO THE SPONSOR, IT MAY BE REFERRED TO AND ANY ADDITIONS MADE TO BRING IT UP-TO-DATE. FORM FDA 1573 (10/82) PREVIOUS EDITIONS ARE OBSOLETE. 105 2a. The investigator assures that an IRB that complies with the requirements set forth in Part 56 of this chapter will be responsible for the initial and continuing review and approval of the proposed clinical study. The investigator also assures that he/she will report to the IRB all changes in the research activity and all unanticipated problems involving risks to human subjects or others, and that he/she will not make any changes in the research that would increase the risks to human subjects without IRB approval. FDA will regard the signing of the Form FDA 1573 as providing the necessary assurances stated above. b. A description of any clinical laboratory facilities that will be used. (If this information has been submitted to the spon- sor and reported by him on Form FDA 1571, reference to the previous submission will be adequate). 2. he investigational drug will be used by the undersigned or under his supervision in accordance with the plan of investi- gation described as follows: (Outline the plan of investigation including approximation of the number of subjects to be treated with the drug and the number to be employed as con- trols, if any, clinical uses to be investigated; characteristics of subjects by age, sex and condition; the kind of clinical obser- vations and laboratory tests to be undertaken prior to, during, and after administration of the drug, the estimated duration of the investigation; and a description or copies of report forms to be used to maintain an adequate record of the obser- vations and test results obtained. This plan may include rea- sonable alternates and variations and should be supplemented or amended when any significant change in direction or scope of the investigation is undertaken.) 4. THE UNDERSIGNED UNDERSTANDS THAT THE FOL- LOWING CONDITIONS, GENERALLY APPLICABLE TO NEW DRUGS FOR INVESTIGATIONAL USE, GOVERN HIS RECEIPTS AND USE OF THIS INVESTIGATIONAL DRUG: a. The sponsor is required to supply the investigator with full information concerning the preclinical investigations that justify clinical trials, together with fully informative material describing any prior investigations and experience and any possible hazards, contraindications, side-effects, and precau- tions to be taken into account in the course of the investiga- tion. b. The investigator is required to maintain adequate records of the disposition of all receipts of the drug, including dates, quantities, and use by subjects, and if the investigation is terminated, suspended, discontinued, or completed, to return to the sponsor any unused supply of the drug. If the investi- gational drug is subject to the Comprehensive Drug Abuse Prevention and Control Act of 1970, adequate precautions must be taken including storage of the investigational drug in a securely locked, substantially constructed cabinet, or other securely locked substantially constructed enclosure, access to which is limited, to prevent theft or diversion of the substance into illegal channels of distribution. c. The investigator is required to prepare and maintain ade- quate and accurate case histories designed to record all obser- vations and other data pertinent to the investigation on each individual treated with the drug or employed as a control in the investigation. d. The investigator is required to furnish his reports to the sponsor of the drug who is responsible for collecting and eval- uating the results obtained by various investigators. The spon- sor is required to present progress reports to the Food and Drug Administration at appropriate intervals not exceeding 1 year. Any adverse effect that may reasonably be regarded as caused by, or probably caused by, the new drug shall be reported to the sponsor promptly, and if the adverse effect is alarming, it shall be reported immediately. An adequate report of the investigation should be furnished to the sponsor shortly after completion of the investigation. e. The investigator shall maintain the records of disposition of the drug and the case histories described above for a period of 2 years following the date a new-drug application is ap- proved for the drug; or if the application is not approved, until 2 years after the investigation is discontinued. Upon the request of a scientifically trained and properly authorized employee of the Department, at reasonable times, the investi- gator will make such records available for inspection and copying. The subjects’ names need not be divulged unless the records of particular individuals require a more detailed study of the cases, or unless there is reason to believe that the rec- ords do not represent actual cases studied, or do not represent actual results obtained. f. The investigator certifies that the drug will be administered only to subjects under his personal supervision or under the supervision of the following investigators responsible to him, and that the drug will not be supplied to any other investi- gator or to any clinic for administration to subjects. g. The investigator certifies that he will inform any subjects including subjects used as controls, or their representatives, that drugs are being used for investigational purposes, and will obtain the consent of the subjects, or their representatives, except where this is not feasible or, in the investigator’s pro- fessional judgment, is contrary to the best interests of the subjects. h. The investigator is required to assure the sponsor that for investigations subject to an institutional review requirement under Part 56 of this chapter the studies will not be initiated until the institutional review board has reviewed and approved the study. (The organization and procedure requirements for such a board as set forth in Part 56 should be explained to the investigator by the sponsor.) Name of Investigator Very truly yours, Address. Telephone ( ) (This form should be supplemented or amended from time to time if new subjects are added or if significant changes are made in the plan of investigation.) 106 New Drug Studies Group Application Guidelines I. Introduction The Division of Cancer Treatment of the National Cancer Institute has established the New Drug Studies Group mechanism in order to provide NCI Investigational New Drugs for study by clinical investigators who are not members of NCI approved Cancer Centers. Since DCT policy restricts eligibility to test its drugs to investiga- tors approved by peer review, either under contract, PO1/ROI holders, cooperative group grantees, or who are staff of NCI approved (CORE) Cancer Centers, this mechanism permits other highly qualified clinical researchers to test NCI drugs. It is anticipated that studies conducted by NDSG will be Phase II trials. Under exceptional circumstances Phase III trials may be conducted (See Section 6 of Investigators Handbook). No Phase I trials will be permitted. II. Application Requirements Investigational groups seeking designation as a New Drug Studies Group should indicate in their appli- cation, according to the following outline: a. The proposed Leader of the Group for evaluation of Investigational New Drugs and his qualifications, with particular reference to experi- ence in conducting clinical trials with investigational anticancer drugs. b. Prior experience of participating investigators with the evaluation of investigational drugs. ¢. Prior experience of the group/insti- tution in conducting clinical cancer research. d. The proposed tumor types to be studied, and evidence that sufficient numbers of patients are available to conduct the proposed studies. . An applicant who is a member of an NCI sponsored Cooperative Group must document that the trials under the NDSG will not conflict with commitments to Cooperative Groups. . If any affiliate investigators or in- stitutions are intended, then there should be a written policy for qualifications and standards for affiliates (see Section 13 of the Investigator Handbook) as well as mechanisms for assuring the qual- ity of clinical research by these affiliates (see Section 16 of the Investigator Handbook). . Procedures for data management and assurance of protocol compliance. . A description of: 1) facilities available for in and out patient treatment, 2) institutional programs relating to cancer, including a description of organizational entities and staff which will be responsible for these studies (eg. divisions, departments, sections, etc.), 3) statistical support for evaluation of the results of protocol studies of Investigational New Drugs, and 4) institutional procedures and facilities for storage, dispensing, handling and account of investi- gational drugs, in accord with NCI policies. i. A description of the Institutional Review Board (as defined by Public Law 93-348, Title 2, Section 212(a) of the Code of Federal Regulations and FD Form 1571, Section 10, Part C). j. A signed FD-1573 for each investigator. 109 110 III. Review of Applications 1. The application will be reviewed by staff of the Cancer Therapy Evaluation Program (CTEP). 2. The review will focus on three broad areas: a. The qualifications of the investi- gators and merits of the proposal, as outlined in the application. b. The demonstrated ability of proposer to perform clinical research and the existence of procedures to assure the quality of work. c. The extent to which the pro- posed NDSG provides DCT with a needed resource (e.g., particular scientific expertise or an unusual patient population) which does not duplicate that which is available through already supported mechanisms. IV. Instructions to Applicants 1. The applicant should submit 5 copies of the application to: Head, Protocol and Information Office Cancer Therapy Evaluation Program Division of Cancer Treatment, NCI Landow Building, Room 4C-33 Bethesda, Maryland 20892 2. Written notification of the receipt of the application will be made. 3. The application will be reviewed by the Cancer Therapy Evaluation Program (CTEP) Staff. V. Responsibilities of NDSG Approved applicants are expected to follow all DCT IND drug procedures as outlined in the Investigator’s Handbook. Please pay particular attention to Section 12. Responsibilities of the Protocol Chairman and Clinical Investigators Letter of Intent Investigational Drug Trial National Cancer Institute Cancer Therapy Evaluation Program Drug Dose Schedule Tumor Type Group or Institution Performance Status of Patients Prior Treatment Status of Patients Experimental Design When Would Trial Begin Estimated Annual Accrual Accrual Documented by Prior Trials (include breakdown by P.S. and prior treatment) Proposed Sample Size Projected Date of Completion of Accrual List Competing Studies in the Selected Tumor Type List Competing Studies for Which the Proposed Patient Population will be Eligible Date Protocol Chairman Group Chairman/Contract PI (If applicable) 10/85 113 Informed Consent Checklist I. Required Elements - Must be present in the informed consent document. IL. III. L Clearly state that the study involves research. State which drug(s), treatment(s) or delivery technique(s) is experimental. Clarify the study purpose(s) in layman’s terms. State the patient’s expected duration of participation in study (e.g., the patient will be treated until there is evidence that therapy is no longer effective). Give a brief description of the procedure(s) to be performed to monitor the patient during study (e.g., X-rays, lab evaluations, etc.). An exhaustive list is not necessary. Give a description of the experimental aspect(s) or new delivery technique(s) of the study. State in specific terms the route of administration of each drug (e.g., IV, oral, continuous infusion, etc.). State estimated time of delivery of each drug or time of procedure (e.g., 5 minutes, 30 minutes, 24 hours, etc.). . State which risks are attributed to specific drug(s) or procedures(s). . Clarify and describe expected benefit(s) to be derived from participation in this study (e.g., tumor shrinkage, quality of life, etc.). . In general terms, discuss alternative treatment(s) to participation in this study (e.g., conventional chemotherapy, irradiation, hormonal therapy, surgery, etc.). . State the extent to which confidentiality of records will be maintained. State that a qualified representative of the FDA may inspect patient/study records. State that a qualified representative of the NCI may inspect patient/study records. State if compensation for study related injury will be provided by the institution or other insurer. State if emergency treatment of injury will or will not be provided by the institution. Provide space in the form or list the name(s) and number(s) of contact person(s) for research related questions. Provide space in the form or list name(s) and number(s) of the contact person(s) (not involved in the research) for patient rights related questions. . Clearly state participation is voluntary. State that refusal to participate will involve no loss of benefits or penalize the patient’s care. State that discontinuation of participation in the study will involve no loss of benefits to which the patient is entitled. Additional Elements - May be appropriate for some studies 1. 2 3. 4. 5. 6. State that unforseeable or unexpected risk(s) may be involved. State the circumstances under which the patient’s participation may be terminated by the investigator without the patient’s consent. State that additional costs may be incurred by the patient’s participation in the study. State the consequences of the patient’s decision to withdraw from the study. State that significant new findings that relate to the patient’s treatment will be discussed with the patient. State the approximate number of patients involved in the study. Suggested Elements I: 2. 3. State that a copy of the informed consent form shall be given to the patient. The form should be written in layman’s terms. Reference to approval by the IRB, NCI or Cooperative Group may be misleading to the patient. 117 NAME OF INSTITUTION Protocol Submission Checklist National Cancer Institute Cancer Therapy Evaluation Program INSTITUTION PROTOCOL #: = NCI PROTOCOL #: PROTOCOL CHAIRMAN: PHONE NUMBER: LIST NCI SUPPLIED DRUGS: Please indicate type of material enclosed and fill in blanks: [J New Protocol [J Revised Protocol [J] Amendment [J Status Notice I IL. IIL. IV. NEW PROTOCOL ENTITLED: [J [J HAVE YOU SUBMITTED A LETTER OF INTENT FOR THIS PROTOCOL? Y N IF YES, LOI # [J [0 ARE YOU ENCLOSING A SIGNED AND DATED IRB APPROVAL (Form 596)? Y N IF NO, EXPLAIN [J [J ARE YOU SUBMITTING THIS PROTOCOL FOR APPROVAL FOR CCOP? Y N [J [J IS THIS PROTOCOL PART OF A ROl OR POI? GRANT # Y N [J [J DO YOU WISH THIS PROTOCOL TO BE CONSIDERED A CONTRACT FUNDED STUDY? Y N CONTRACT # PROJECTED DATE OF ACTIVATION. (If already opened, provide activation date ___ ~~) REVISIONS: RESPONSE TO CTEP QUESTIONS FOR PROTOCOL AWAITING CTEP APPROVAL AMENDMENT TO APPROVED PROTOCOL: [J] ACTIVATION AMENDMENT (List of changes between CTEP approval & local activation) [J Activation Amendment Only (list changes) [J Activation Amendment plus replacement pages [J Activation Amendment plus replacement document [Ll AMENDMENT FOR ACTIVE STUDY [J] Editorial, Administrative changes [J] Change of participants [J Scientific changes [J Change of Protocol Chairman OFFICIAL NOTICE OF CHANGE IN PROTOCOL STATUS: [J] ACTIVATION Date: [J TEMPORARY CLOSURE Date: [] REACTIVATION Date: [J] CLOSURE Date: [J COMPLETION Date: OTHER: SIGNATURE OF PERSON COMPLETING THE FORM/TELEPHONE NUMBER/DATE 7/85 121 Group Protocol Submission Checklist National Cancer Institute Cancer Therapy Evaluation Program NAME OF GROUP: GROUP PROTOCOL #: ~~ NCI PROTOCOL # (if different): PROTOCOL CHAIRMAN: PHONE #: LIST NCI SUPPLIED DRUGS: Please indicate type of material enclosed and fill in blanks: [] New Protocol [] Revised Protocol [J Amendment [] Status Notice I. NEW PROTOCOL ENTITLED: [J] [J HAVE YOU SUBMITTED A LETTER OF INTENT OR A CONCEPT REVIEW FOR THIS Y PROTOCOL? IFYES, Lo1#__ = orCR# [J [J IS THIS A PILOT STUDY SUBMITTED FOR “INFORMATION & FILING ONLY” (No IND Y N drugs and <100 patients)? [J [J DOES THIS PROTOCOL INCLUDE A NEW MASTER PROTOCOL? Y N (J [J IS THIS AN INTERGROUP STUDY? Y N (J [J WILL THIS PROTOCOL BE OPEN TO ANY CCOP(S)? Y N PROJECTED DATE OF ACTIVATION (Month/Year) II. REVISIONS: RESPONSE TO CTEP QUESTIONS FOR PROTOCOL AWAITING CTEP APPROVAL III. AMENDMENT TO APPROVED PROTOCOL: [] REQUEST FOR PRIOR APPROVAL [J ACTIVATION AMENDMENT (List of changes between CTEP approval & Group activation) [J Activation Amendment Only (list of changes) [J Activation Amendment plus replacement pages [J Activation Amendment plus replacement document [J AMENDMENT FOR ACTIVE STUDY [J Editorial, administrative changes [J Change of participants [J Scientific changes [J Change of Protocol Chairman IV. OFFICIAL NOTICE OF CHANGE IN PROTOCOL STATUS: [J ACTIVATION Date: [J TEMPORARY CLOSURE Date: [J REACTIVATION Date: [J CLOSURE Date: [J COMPLETION Date: V. OTHER: SIGNATURE OF PERSON COMPLETING THE FORM/TELEPHONE NUMBER/DATE 7/86 122 CTEP Glossary ACTIVATION: The decision by a Group/Institution to open an IND study for patient entry (which occurs after CTEP approval). ACTIVATION AMENDMENT: Any protocol change which occurs after CTEP approval and prior to local activation. Examples: the study is approved by CTEP with recommen- dations which are incorporated prior to activation; these changes must be listed and submitted to CTEP as an Activation Amendment. ADR: Adverse Drug Reaction - An ‘alarming’ ADR is any serious, fatal, or life-threatening clinical experience in a patient which is thought to be drug related. It must be reported immediately to the drug sponsor. “Other” ADR’s are reported if that effect has not been described previously. AMENDMENT: Any protocol change which occurs after activation. APPROVAL: CTEP approves the protocol in writing when the science and informed consent are acceptable, the IRB documentation is on file (not applicable to Groups), and the drugs to be supplied are specified by the Drug Management and Authorization Section. If recommendations are specified, CTEP expects an ‘‘Activa- tion Amendment’’ to indicate any changes to the approved document. BES: Biologics Evaluation Section, IDB, CTEP, DCT, NCI. BRB: Biometric Research Branch, CTEP, DCT, NCI. BRMP: Biological Response Modifiers Program, DCT, NCI. CANCER CENTER: An institution which is designated by NCI as a comprehensive or clinical cancer center and is eligible to conduct IND drug studies. CCOP: Community Clinical Oncology Program - CCOP is a cooperative agreement supported program which provides support to community-based oncologists to participate in clinical trials sponsored by the clinical cooperative groups and/or cancer centers. Each CCOP is expected to enter a minimum of 50 patients per year on NCl-approved research protocols. CIB: Clinical Investigations Branch, CTEP, DCT, NCI CLINICAL BROCHURE: This document contains all relevant information about the drug, including animal screening, preclinical toxi- cology, and detailed pharmaceutical data. Also included, if available is a summary of current knowledge about pharmacology and mechanism of action and a full description of the clinical toxicities. CLINICAL COOPERATIVE GROUPS: Cancer clinical cooperative groups are composed of investigators who join together to develop and implement common protocols. The distinguishing characteristic of cooperative groups is the central operations and statistical offices which support the administrative require- ments of the research and perform central data collection and analysis. CLINICAL TRIALS MONITORING SERVICE: An organization which receives, reviews, and performs data management tasks on individual patient case report forms for Phase I and some Phase II NCI investiga- tional drug studies. 125 126 CLOSED: The decision by a Group, Institution, or NCI to close a study to new patient entries; previously entered patients will continue treatment. COMPLETED: The study is closed and no patients are being treated or followed for data collection. COOPERATIVE ONCOLOGY GROUP ASSURANCE. An agreement for the protection of human research subjects filed with the OPRR by an institution participating in cooperative group trials. CTEP: Cancer Therapy Evaluation Program, DCT, NCI. CTEP LETTER: Newsletter which announces the approval of new drugs for clinical trials and other drug development information. DCPC: Division of Cancer Prevention and Control, NCI. DCS: Developmental Chemotherapy Section, IDB, CTEP, NCI. DCT: Division of Cancer Treatment, NCI. DHHS: Department of Health and Human Services. DRUG ACCOUNTABILITY RECORD FORM: Form used to maintain records of disposition of NCI investigational drugs. NIH Form-2564. DMAS: Drug Management and Authorization Section, IDB, CTEP. DRAS: Drug Regulatory Affairs Section, RAB, CTEP, DCT, NCI. DRUG MONITOR: A physician in IDB is assigned to each IND drug to coordinate its clinical development. DTP: Developmental Therapeutics Program, DCT, NCI. FDA: Food and Drug Administration, DHHS. FDA-1573: Also referred to as a “Statement of Investigator;” it is a requirement of section 505(i) of the Food, Drug, and Cosmetic Act and {312.1 of Title 21 CFR, that an investigator complete this form as a condition for receiving and conducting clinical studies involving investiga- tional drug(s). It includes the investigator’s training and experience and provides for legal certifications. FORM NIH-986: Clinical Drug Request Form. IDB: Investigational Drug Branch, CTEP, DCT, NCI IND: Investigational New Drug Appli- cation - The IND is the legal mechanism under which experimental drug research is performed in the United States. An IND is submitted to the Food and Drug Administration in order to receive an exception from premarketing approval requirements so that experimental clinical trials may be conducted. INVESTIGATOR: Any physician who assumes full responsibility for the treatment and evaluation of patients on research protocols as well as the integrity of the research data. LOI: The Letter of Intent is an in- vestigator’s declaration of interest in conducting a Phase II trial with a specific investigational drug in a particular disease. Approval of the LOI by CTEP commits an investigator to submit a protocol within a specified timeframe. MULTIPLE PROJECT ASSURANCE (MPA): Is a formal written agreement with the Office of Protection from Research Risks (on behalf of the Secretary of DHHS) and an institution which conducts or supports a large amount of DHHS sponsored research involving human subjects, the MPA specifies how the institution will implement the DHHS regulations at 45 CFR 46. NCI: National Cancer Institute, NIH, DHHS. NDA: New Drug Application is the formal process by which the FDA makes the drug generally available to patients and physicians for specific medications. NEW DRUG STUDY GROUP: Highly qualified clinical researchers at an institution specifically approved by IDB to participate in NCI’s drug development program. NIH: National Institutes of Health, DHHS. OPRR: Office for Protection from Research Risks, NIH. OFFICIALLY FILED: At the time of CTEP approval, the protocol docu- ment, the informed consent, or amendment is placed in the “approved” PIO file and is distrib- uted to the Drug Management and Authorization Section, the Clinical Trials Monitoring Services, the Food and Drug Administration, and/or PDQ. PDQ: The Physician Data Query is an on-line data base which makes state- of-the-art treatment information, directory information, and protocol information available to primary care physicians. This data base is main- tained by the International Cancer Research Data Base Branch, International Cancer Information Center, NCI. PIO: The Protocol and Information Office, CTEP, DCT manages the protocol and amendment review process and maintains the official record of all NCI sponsored protocols as well as voluntary protocols for PDQ. PHS: Public Health Service, DHHS. PRB: Pharmaceutical Resources Branch, DTP, DCT, NCI, NIH, DHHS. PRC: The CTEP Protocol Review Committee reviews and approves all studies involving DCT investigational drugs, cooperative group, or CCOP credit. PRINCIPAL INVESTIGATOR (PI): Name of physician who has organiza- tional and fiscal responsibility for the use of federal funds to conduct a plan of research which frequently includes several clinical trials, i.., Contract PI, Group Chairman, ROI or POI, PI, etc. PROTOCOL CHAIRMAN: The scientific coordinator of the study who is responsible for developing and monitoring the study as well as analyzing, reporting, and publishing its results. QACS: Quality Assurance and Compliance Section, RAB, CTEP, DCT, NCI. 127 128 QUALITY ASSURANCE: The monitoring of a clinical trial to assure the quality of the data that supports scientific conclusions. RAB: Regulatory Affairs Branch, CTEP, DCT, NCI, NIH. RESEARCH BASE: An institution or cooperative group which assumes a broad range of responsibilities and functions for the support of clinical trials conducted under its name. It supports the investigator in develop- ing, organizing, implementing, and analyzing clinical trials. It assumes responsibility for the quality of the research, both in concept and execu- tion, and has an important role in assuring patient safety. REVISIONS: Any protocol change which occurs between initial submis- sion and CTEP approval and official filing. SINGLE PROJECT ASSURANCE (SPA): Is a formal written agreement with the Office of Protection from Research (on behalf of the Secretary of DHHS) and an institution which has a Multiple Project Assurance and conducts a DHHS - sponsored research project. The SPA specifies how the institution will implement the DHHS regulations a 45 CFR 46. SPONSOR: An organization or indi- vidual who assumes legal responsi- bility for supervising or overseeing clinical trials with investigational agents. TEMPORARILY CLOSED: The decision by a Group, Institution, or NCI to stop patient entry pending study evaluation. I€1 "U.S. GOVERNMENT PRINTING OFF ICE: 1984—446-045 NIH-986 REV. 6-84 The drugs listed below are requested for the use of: FOR NCI USE ONLY RETURN COMPLETED FORM TO: ORDER NUMBER oa7E NATIONAL INSTITUTES OF HEALTH Drug Management and Authorization Dr. oo No. NATIONAL CANCER INSTITUTE gation Caner Fraalment, NC) Requester (if other than investigator): SIGNATURE OF AUTHORIZING OFFICIAL Landow Building, Room 222 CLINICAL DRUG REQUEST 7910 Woodmont Avenue oo Bethesda, Maryland 20205, U.S.A. me Title: Tel. STRENGTH & QUANTITY PROTOCOL NO. YOUR NSC DOSE FORM ORDERED MANUFACTURER OR IND PT. CURRENT DRUG NAME NUMBER (Specify vials, tablets, (Specity vials & LOT NUMBER COMMENTS NUMBER * INVENTORY capsules) or bottles) A B C D E F G H INSTRUCTIONS ‘Number of patients currently on study. IPPING INFORMATION 1. Dre ALL INFORMATION — One item or protocol per P ™ line. Investigator Signature Date: DATE SHIPPED: 2 fir in all sections completely including the shipping address. 3. DO NOT cross the double line into the shaded area Operations Office/ Research Base Verification Date marked FOR NCI USE ONLY. How Shipped: Pulled ty: 4. Limit drug request to an eight (8) week supply : 5. Sign and date the order y 1. [J Room Temperature 6. Return the completed form to the address indicated: PLEASE SPECIFY AFFILIATION BELOW: 2. [J Refrigerate On above. Ark 1. [J Study Group 6. [J International Agreement al . : In 7 Shipping Address Must Be Typed in Space Below. 7. [J Group C (Guidelines) 3. [J Packed In Dry Ice Checked by: 4. [7] Priority Mail 2. [J Contract 8. [J ccop 5. [J GBL 3. [J Phase | Working Group 6. [J Other (Specify) 4. [J Cancer Center 9. [] Special Exception Packaged by, 10. [J Nonclinical Use 5. [J NCI Intramural 11. [J Other (Specify) 1. PHARMACEUTICAL RESOURCES BRANCH ERVIC, NS Es vs \ ed A» Ty DEPARTMENT OF HEALTH & HUMAN SERVICES Public Health Service National Institutes of Health National Cancer Institute Bethesda, Maryland 20892 Revised Guidelines Reporting of Adverse Drug Reactions October 1985 TO : Clinical Investigators FROM: Investigational Drug Branch, CTEP, DCT, NCI Enclosed are revised guidelines for the reporting of adverse drug reactions (ADRS) in patients treated with investigational agents supplied under an IND sponsored by the Division of Cancer Treatment, NCI. The timely reporting of ADRs is required by the Food and Drug Administration (FDA) regulations and such reporting improves both patient care and scientific communication by allowing the NCI to rapidly disseminate the new findings to the investigator community studying the drug. The requirement for timely reporting is clearly stated in the investigator registration form FD-1573 (see Attachment I). In signing the FD-1573 you accept the responsibility for conducting investigational drug trials which includes reporting ADRs to the Division of Cancer Treatment, NCI. Please note that changes and clarification in the reporting requirements have been made for Phase I studies as well as for Phase II and III studies. In addition, a new form for reporting ADRs to NCI is included. The additional attachments include the following: Attachment 2: An outline and discussion of the requirements for reporting ADRs Attachment 3: NCI form for reporting ADRs occurring with investigational agents Attachment 4: FDA Form 1639 to be used to report ADRs occurring with commercial agents. 135 Attachment 1 October 1985 Adverse Drug Reactions FDA Regulations NCI RESPONSIBILITY “The sponsor shall promptly investigate and report to the FDA and to all investigators any findings associated with the use of the drug that may suggest significant hazards, contraindications, side effects and precautions pertaining to the safety of the drug. If the finding is alarming, it shall be reported immediately and the clinical investigation discontinued until the finding is adequately evaluated and a decision is reached that it is safe to proceed”. Form FD-1571 INVESTIGATOR RESPONSIBILITY “‘Any adverse effect that may reasonably be regarded as caused by, or probably caused by, the new drug shall be reported to the spon- sor promptly, and, if alarming, it shall be reported immediately’. Form FD-1573 136 Attachment 2 October 1985 NCI Division of Cancer Treatment Adverse Drug Reactions with Investigational Agents RESPONSIBILITIES OF INVESTIGATORS The prompt reporting of adverse drug reactions (ADRs) to DCT is the responsibility of each investiga- tor engaged in clinical research with investigational drugs supplied by NCI. It is the policy of the DCT that investigators are encouraged to submit such reports even if there is only a suspicion of a drug effect. Procedures for reporting ADRs are outlined in the attached chart. In all instances investigators should be prepared to provide additional information to NCI if requested. Any investigator who is dubious about whether a particular adverse reaction needs to be reported should call the IDB at 301-496-7957. Failure to report adverse reactions in a timely manner may result in discontination of the study, and, in some cases, revocation or suspen- sion of the investigator’s permission to perform clinical research using investigational drugs under the Division of Cancer Treatment, NCI sponsored IND. This reporting of adverse reactions is in addition to and does not supplant the reporting of toxicities as part of the report of the results of the research protocol, e.g., cooperative group minutes. All adverse reactions should also be reported to your local Institutional Review Board. Adverse Drug Reactions Occurring on Phase I Trials Life-threatening reactions which may be due to drug administration and all fatal reactions occurring on Phase I studies should be reported immediately by phone. A written report should be submitted within ten (10) working days. The first occurrence of any toxicity, regard- less of grade, should be reported by phone within 24 hours to the appropriate Drug Monitor in IDB. An ADR report may be required. Adverse Drug Reactions Occuring on Cooperative Group Studies Investigators on protocols of Cooperative Groups should check with the Operations Office and/or the Chairman of the New Agents Committee about their practices for handling these reports, which may be routed through them if a mechanism is in place to do that on an emergency basis. Adverse Drug Reactions Occurring with Commercial Drugs These toxicities should be reported promptly in writing (ATTACH- MENT 5, Form 1639) if that type of effect has not previously been reported in the package insert and/or the literature. A clear description of the suspected reaction should be provided along with an assessment as to whether the event is drug- or disease-related. The report should be sent to the IDB within ten (10) working days of its occurrence. Attachment 2 (continued) Investigational Agent Adverse Drug Reaction Reporting Chart National Cancer Institute, Division of Cancer Treatment Phase 1 Studies Reaction Reporting Obligation a. ALL LIFETHREATENING EVENTS REPORT BY PHONE TO IDB WITHIN (Grade 4)> WHICH MAY BE DUE TO 24 HOURS.! DRUG ADMINISTRATION. b. ALL FATAL EVENTS. REPORT BY PHONE TO IDB WITHIN 24 HOURS.! (written report to follow within 10 working days?) ¢. FIRST OCCURRENCE OF ANY TOXICITY. REPORT TO THE DRUG MONITOR WITHIN 24 (regardless of grade) HOURS. (an ADR report may be required) Phase II and III Studies UNKNOWN REACTION*? KNOWN REACTION*? GRADES 2-32 GRADES 4 AND 5 GRADES 1-3 GRADES 4 AND 5 Written report Report by phone to Not to be reported Written Report to to IDB within IDB within 24 hrs.!. as ADRs. These IDB within 10 10 working days. Written report to toxicities should be working days. follow within 10 be submitted as (grade 4 myelo- working days. part of study suppression not to summary. be reported.) ‘“‘Aplasia in leu- kemia patients®”’ USE ATTACHMENT 4 TO REPORT ADRs WITH DCT SPONSORED DRUGS. Telephone number available 24 hours daily: 301-496-7957 Use protocol designated toxicity grading scale. 3Report to: Investigational Drug Branch P.O. Box 30012 Bethesda, Maryland 20814. 4A list of all known toxicities can be found in the protocol or informed consent form of the protocol. Reactions definitely felt not to be treatment related should not be reported. However, a report should be submitted if there is only a suspicion of drug effect. Grade 4 unknown and known reactions not to be reported. Report Grade 5 unknown and known reactions within 10 working days if clearly related to experimental drug (i.., single agent therapy). Otherwise do not report. 137 138 Attachment 3 ADR # (Assigned at NCI) NCI Adverse Reaction Form for Investigational Agents Person Completing this Form Date Data Supplied by Phone Call Other (For use by NCI) (Specify) I. DEMOGRAPHICS A. Patient Information PT LD. # Age Sex Date of Initial DX Malignancy Site of Primary PS (At start of study) Site(s) of Metastatic Disease Concurrent Non-Malignant Disease and Non-Protocol Medications B. Drug Information Drug Name Source of Drug: NCI ___ ~~ Other _____ (specify ) Type of Reaction Toxicity Grade Date of Reaction Date IRB Notified NCI Protocol # Investigator Phase of Study Institution Phone ( ) Protocol Treatment (include all agents) Drug Dose Schedule Route Date of First Course Number of Courses Date Last Course Started __ ~~ Date of Therapy Associated with ADR Prior Therapy (Drug, radiation, relevant surgery: Include dates of therapy) II. DOCUMENTATION OF REACTION A. Non-Myelosuppressive Toxicity and Previously Unknown Myelosuppression 1. Description of Reaction and Temporal Relationship to Investigational Drug Administration 2. Physical Findings and Laboratory Data Documenting Toxicity 3. Treatment of Adverse Reaction 139 4. Complications and Sequelae (If death, was an autopsy obtained?) Attachment 3 (continued) 5. Past History of Organ Dysfunction 6. Rechallenge with Agent _ No __ Yes If yes, describe: Myelosuppression (Previously known or unknown) 1. Laboratory Data Documenting Myelosuppression Recovery or Baseline Nadir Latest Value Date/ Value Date/ Value Date/ Value WBC or PMN / / / Platelets / / / Hgb or Hct / / 7 2. Complications, Treatment and Sequelae (e.g. infections/hemorrhage) Grade of Toxicity and Reporting Requirements (Check one) 1. Previously Unknown Toxicities: a. Fatal or Life Threatening __ (Report by Telephone in 24 hours: 301-496-7957) Date _____ NCI Contact b. Grade I II 11 (Send form within 10 days) 2. Previously Known Non-Myelosuppressive Toxicities: a. Fatal or Life Threatening (Send form within 10 days) 3. Previously Known Myelosuppressive Toxicities: a. Fatal (Send form within 10 days) Send Forms to: Investigational Drug Branch, NCI P.O. Box 30012 Bethesda, Maryland 20814 Investigator’s Assessment (If more than 1 investigational agent was used, give an assessment for each by writing the drug names on the appropriate lines) IND Non-IND Drug Drug Disease Action Taken: Therapy Required: Unrelated None None Unlikely Dose Reduced __ Symptomatic Possible Dose Withheld Supportive Probable Drug Discontinued __ Intensive Definite N/A E. Additional narrative may be sent on separate page if required 140 Attachment 4 EPARTMENT OF HEALTH AND HUMAN SERVICES PUBLIC HEALTH SERVICE FOOD AND DRUG ADMINISTRATION (HFN-730) ROCKVILLE, MD 20857 ADVERSE REACTION REPORT (Drugs and Biologics) Form Approved: OMB No. 0910-0230. FDA CONTROL NO. ACCESSION NO. REACTION INFORMATION PATIENT ID/INITIALS (In Confidence) 2. AGE 3. SEX 4.-6. REACTION ONSET YRS. MO. DA. YR. DESCRIBE REACTION(S) [] RELEVANT TESTS/LABORATORY DATA OJ 8.-12. CHECK ALL APPROPRIATE: PATIENT DIED REACTION TREATED WITH Rx DRUG RESULTED IN, OR PROLONGED, INPATIENT HOSPITALIZATION RESULTED IN PERMANENT DISABILITY NONE OF THE ABOVE SUSPECT DRUG(S) INFORMATION SUSPECT DRUG(S) (Give manufacturer and lot no. for vaccines/biologics) . DAILY DOSE 16 ROUTE OF ADMINISTRATION 20. DID REACTION ABATE AFTER STOPPING DRUG? [] ves (Jno [OO na . INDICATION(S) FOR USE . DATES OF ADMINISTRATION (From/To) 19 DURATION OF ADMINISTRATION J YES 21. DID REACTION REAPPEAR AFTER REINTRODUCTION? [Aw [J VW CONCOMITANT DRUGS AND HISTORY _ CONCOMITANT DRUGS AND DATES OF ADMINISTRATION (Exclude those used to treat reaction) _ OTHER RELEVANT HISTORY (e.g. diagnoses, allergies, pregnancy with LMP, etc.) ONLY FOR REPORTS SUBMITTED BY MANUFACTURER V. INITIAL REPORTER (In confidence) . NAME AND ADDRESS OF MANUFACTURER (Include Zip Code) 26.-26a. NAME AND ADDRESS OF REPORTER (Include Zip Code) a. IND/NDA. NO. FOR SUSPECT DRUG 24b MFR. CONTROL NO. 26b. TELEPHONE NO. (Include area code) c. DATE RECEIVED BY MANUFACTURER 24d. REPORT SOURCE (Check all that apply) [Jroreicn [] sTuov [JuTERATURE [[JHeALTH PROFESSIONAL [Jconsumer [Ino 26¢. HAVE YOU ALSO REPORTED THIS REACTION TO THE MANUFACTURER? [] ves i 15 DAY REPORT? Clves [no 25a. REPORT TYPE [1] wimac [J rorcowue 26d. ARE YOU AHEALTH PROFESSIONAL? C1 ves [no OTE: Required of manufacturers by 21 CFR 314.80 Submission of a report does not necessarily constitute an admission that the drug caused the adverse reaction. IM FDA 1639 G 86) PREVIOUS EDITION MAY BE USED 141 INSTRUCTIONS FOR COMPLETING FORM FDA 1639 Attachment 4 (continued) !Jse a separate report form for each case. If more space is needed, additional pages may be attached. 1s ® NOOO, WN 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 142 I. Patient/Reaction Information (Items 1-12) Patient ID/Initials: Record patient's identification (i.e. medical record number, initials, etc). (This information is kept in confi- dence by the FDA.) . Age: Racord the age of the patient. When reporting a congenital malformation, record the age of the mother. . Sex: Record the sex of the patient. When reporting a congenital malformation, record the sex of the baby. . Weight: Record the weight of the patient in pounds. When reporting a congenital malformation, record the weight of t! : mother. . Height: Record the height of the patient in inches. When reporting a congenital malformation, record the height of the mother. . Reporting Date: Record the date when the report was initially communicated to the manufacturer. . Reacticn Onset Date: Record the date on which the reaction was first observed or detected. . Suspected Reaction(s): Describe the signs, symptoms and course of the drug related event in the terminology used by the original observer of the reaction. (Coding terms e.g. COSTART, SNOMED, etc. may also be noted, but only in addition to original description.) . Outcome of Reaction: Indicate the status of the patient as of date indicated in Item 23. If the patient died, give the cause and date of death. Include discharge summary and/or autopsy findings, if available. Tests/l.aboratory Data: Describe the results of all diagnostic tests and exams (e.g. biochemical tests, x-rays, endoscopy, biopsy, etc.) which were done as a result of the event described in Item 8. Pertinent base line values and laboratory normals should be included with each test or exam reported. If this information is not available at the time of the initial report, a follow up report should be submitted. Treatn.ent Required: If “yes”, a short description of treatment should be included in Item 8. Hospitalization Required: If ““yes’’, a short description of the treatment should be included in Item 8. Il. Suspect Drug Information (Items 13-20) Suspect Drug(s): Record the trade name. The generic name should be used only when the trade name is not known. Include IND/NDA number of the drug as well as the lot number, when available. Total Daily Dose: Record the total daily dose as of the date recorded in Item 7. If drug(s) was given in a different dose or form oi a previous occasion, include dates and total daily dose for each drug exposure. Route of Administration: Record the route of administration (i.e. po, IM, IV) as of the date recorded in Item 7. Indication(s) for use: Record intended use in accepted medical terminology. Therapy Dates: Give starting and stopping dates of administration for each drug listed in Item 13. Therapy Duration: Give duration of therapy in days. Dechallenge: (a) Applicable if the suspect drug(s) was either reduced in dosage or discontinued. (b) If 19(a) is checked, indicate whether the reaction subsided upon reduced dosage or discontinuation of the drug. Rechallenge: (a) Applicable if the suspect drug was reintroduced to the patient's therapy after dechallenge. (b) If 20(a) is “'yes”, indicate whether or not the reaction reappeared upon rechallenge with the drug. Il. Recent/Concomitant Drugs and Medical Problems (Items 21-22) List all recent or concomitant drugs. Include the total daily dose(s), indication(s) for use, route(s) of administration and dates of administration and/or duration of therapy for each drug. Describe other relevant medical conditions or problems which could have contributed to the reaction. Include pertinent medical history such as allergies, occupation, industrial hazards, diet, smoking, climate, ethnic origin, cosmetics and biologicals. When re- porting a congenital malformation, include the date of the last menstrual period of the mother, gravidity, parity and previous abortions. IV. Other Information (Items 23-26) Manufacturers Information: Include manufacturer's name, address, control number and date report is sent to FDA. This control number is the identifying number assigned by the manufacturer to the report for internal record control. Indicate if this is an initial submission to FDA or a follow-up of a previously submitted Form FDA 1639. If this is a follow-up attach copy of initial report. Record the name, title and address of the practitioner originating the report. (This information is kept in confidence by the FDA.) Check “yes or “no”, if the source of this report may or may not be released to the Armed Forces Institute of Pathology for further study and follow-up. This is encouraged whenever possible. OS oo A Ee BE rr ® — a oo Ey 9 gd . OMB NO. 0925-0240 EXPIRES: 9/30/87 National Institutes of Health National Cancer Institute Investigational Drug Accountability Record PAGE NO. CONTROL RECORD O SATELLITE RECORD O Name of Institution Protocol No. (NCI) Drug Name, Dose Form and Strength Protocol Title Dispensing Area Investigator Date Line Patient's No. Initials Patient's |.D. Number Dose Quantity Dispensed or Received Balance Forward Balance Recorder's Initials Manufacturer and Lot No. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21, 22. 23. 24. NIH-2564 9-85 145 FE National Study Recommendations Commission on for Handling Cytotoxic Exposure Cytotoxic Agents Preamble The increasing use of cytotoxic agents and the growing awareness of potential hazards requires special attention to the procedures utilized in the handling, preparation and administration of these drugs. Equally important is the proper disposal of chemical residues and wastes. These recommendations are intended to provide information for the protection of personnel participating in the clinical process of chemotherapy. The mutagenic and carcinogenic potential of many cytotoxic agents is well established and is a possible hazard to the health of exposed individuals. It is the responsibility of institutional and private health care providers to adopt and use appropriate procedures for protection and safety. |. Environmental Protection 1. All mixing of cytotoxic agents should be performed in a Class II, biological safety cabinet. Type A cabinets are the minimal requirement. Type A cabinets which are vented (some now classified as Type B3) are preferred. 2. Special techniques and precautions must be utilized because of the vertical (down- ward) laminar airflow (see Supplement I). 3. The biological safety cabinet must be certified by qualified personnel annually or any time the cabinet is physically moved. 4. The biological safety cabinet should be operated with the blower on, 24 hours per day — seven days per week. 5. Drug preparations must be performed only with the view screen at the recommended access opening. Professionally accepted practices concerning the aseptic preparation of injectable products should be followed. Il. Operator Protection 1. Disposable surgical latex gloves are recommended for all procedures involving cytotoxic drugs. Polyvinyl chloride (PVC) gloves should not be worn while handling cytotoxic agents. Several types of PVC gloves are permeable to a variety of drugs. 2. Gloves should routinely be changed approximately every 30 minutes when working steadily with cytotoxic agents. Gloves should be removed immediately after overt contamination. 3. Double gloving is reccommended for cleaning up of spills. 4. Protective barrier garments should be wom for all procedures involving the prepara- tion and disposal of cytotoxic agents. These garments should have a closed front, long sleeves and closed cuff (either elastic or knit). wn . All potentially contaminated garments must not be worn outside the work area. lll. Compounding Procedures 1. Hands must be washed thoroughly before gloving and after gloves are removed. and Techniques . . ; ; : q _ Care must be taken to avoid puncturing of gloves and possible self-inoculation. . Syringes and LV. sets with Luer-lock fittings should be used whenever possible. HH Wo _ Vials should be vented with a hydrophobic filter to eliminate internal pressure or vacuum. wn . Before opening ampules, care should be taken to insure that no liquid remains in the tip of the ampule. A sterile, disposable alcohol dampened gauze sponge should be wrapped around the neck of the ampule to reduce aerosolization. 6. For sealed vials, final drug measurement should be performed prior to removing the needle from the stopper of the vial and after the pressure has been equalized. 7. A closed collection vessel should be available in the biological safety cabinet or the original vial may be used to hold discarded excess drug solutions. 8. Special procedures should be followed for acute exposure or spills (Supplement II). 9. Cytotoxic agents which are handled within the treatment area should be properly labeled (e.g. “Chemotherapy: Dispose of Properly”). PR ST ST LS Er Approved by the National Study Commission on Cytotoxic Exposure March 1984 149 IV. Precautions for Medication Administration » V. Disposal Procedures VI. Personnel Policy Recommendations VII. Monitoring Procedures No Appendix XIII (continued) - Disposable surgical latex gloves should be worn during all cytotoxic drug administra- tion activities. - Syringes and LV. sets with Luer-lock fittings should be used whenever possible. - Special care must be taken in priming LV. sets. The distal tip cover must be removed before priming. Priming should be performed into a sterile, alcohol-dampened gauze sponge, which then is disposed of appropriately. . Place contaminated materials in a leakproof, puncture-proof container appropriately marked as hazardous waste. - Cytotoxic drug waste should be transported according to the institutional procedures for contaminated material. - There is insufficient information to recommend any single preferred method for disposal of cytotoxic drug waste. 3.1 One method for disposal of hazardous waste is by incineration at a temperature considered sufficient by the Environmental Protection Agency (EPA) to destroy organic compounds. Incineration should be done in an EPA permitted hazardous waste incinerator. 3.2 Another method of disposal is by burial at an EPA permitted hazardous waste site. 3.3 A licensed hazardous waste disposal company may be consulted for information concerning available methods of disposal in the local area. - All personnel working with cytotoxic agents must receive special training. - Access to the compounding area must be limited to only necessary authorized personnel. . The personnel working with these agents should be observed regularly by supervisory personnel to insure compliance with procedures. - Acute exposure episodes must be documented. The employee must be referred for professional medical examination. - Procedures to monitor the equipment and operating techniques of the personnel should be performed on a regular basis and documented. Specific methods of monitoring should be developed to meet the complexities of the function. - Itisrecommended that personnel involved in the preparation of cytotoxic agents on a full time basis be given periodic health examinations in accordance with institutional policy. Supplement | 150 Special Techniques and Precautions for Use In the Class II Biological Safety Cabinet - All equipment needed to complete the procedure in the Class IT Biological Safety Cabinet should be placed into the cabinet before beginning and the view screen should be placed at the recommended operating position. A wait of at least two to three minutes before beginning work to allow the unit time to purge itself of airborne contaminants is recommended. . The proper procedures for use in the Biological Safety Cabinet are not the same as those used in the horizontal laminar hood. In many cases they seem contradictory, although in theory they are not. This is because of the nature of the airflow pattern in the Biological Safety Cabinet. Clean air descends through the work zone from the top of the cabinet toward the work surface. As it descends, the air is split, with some leaving through the rear perforation and some leaving through the front perforation. 9. 10. Supplement Il Appendix XIII (continued) The region where the airflow splits is known as the “smoke split” because smoke introduced into this area appears to split into two directions. . It is recommended that the smoke split be determined and marked on each cabinet after it is purchased even if the manufacturer states its location. This can be easily done by using an incense stick to generate smoke and moving it gently from front to rear laterally along the work surface of the cabinet near the center. . Routinely used large equipment should be placed in the cabinet in its normal position when the determination of the smoke split is made. The equipment should then be placed in the same position every time the cabinet is used. . Personnel should refrain from applying any face powder, eye make-up, rouge, finger- nail polish, hairspray or other cosmetics in the work area. These cosmetics may provide a source of prolonged exposure if contaminated. . Eating, drinking, chewing of gum, storage of food or smoking in, around or near the Biological Safety Cabinet should be prohibited. Each of these are sources of ingestion if they are accidentally contaminated by the cytotoxic agent or other hazardous products. . Sterile products should be arranged in the cabinet so as to minimize the possibility of contamination. This may mean locating them in the immediate vicinity of the smoke split. If appropriate, due to quantity or configuration, the sterile items should be kept only in the center and nonsterile items on either side. . For additional operator protection, it is reccommended that the area behind the smoke split be used whenever possible since the airflow direction in that area is away from the operator, lessening the chance of accidental exposure. The least efficient area of the cabinet in terms of product and personnel protection is within three inches of the sides near the front opening. Therefore, you should not work within three inches of the sides of the cabinet. Periodic evaluation of the smoke split should be performed on a routine basis. A constantly changing smoke split location may be indicative of problems with the operation of the cabinet. . Entry into and exit from the cabinet should be in a direct manner perpendicular to the face of the cabinet. Rapid movements of the hands in the cabinet and laterally through the protective air barrier should be avoided. Special Procedures for Acute Exposure or Spills ACUTE EXPOSURE 1.1 Overtly contaminated gloves or outer garments should be removed and replaced immediately after an exposure. 1.2 Hands should be washed after removing gloves. Gloves are not a substitute for handwashing. 1.3 In case of skin contact with a cytotoxic drug product, the affected area should be washed thoroughly with soap and water as soon as possible. Refer to professional medical attention as soon as possible. 1.4 For eye exposure, flush affected eye with copious amounts of water. Refer to professional medical attention immediately. 2.0 SPILLS 2.1 All personnel involved in the clean-up of a spill should wear protective clothing (e.g. gloves, gowns, etc.). All clothes and other material used in the process should be treated or disposed of properly. 2.2 Double gloving should be used in the cleaning up of spills. 151 Appendix XIII (continued) National Study Commission on Cytotoxic Exposure Louis P. Jeffrey, Sc.D., Chairman Director of Pharmacy Services Rhode Island Hospital Roger W. Anderson, M.S. Director of Pharmacy University of Texas M.D. Anderson Hospital Clarence L. Fortner, M.S. Head, Drug Management and Authorization Section IDB Cancer Therapy Evaluation Program Division of Cancer Treatment National Cancer Institute Joseph F. Gallelli, Ph.D. Chief, Pharmacy Department The Clinical Center National Institutes of Health Dennis M. Hoffman, Pharm.D. Director of Pharmacy Services University of New York at Stony Brook For additional information contact: Louis P. Jeffrey, Sc.D., Chairman Louis A. Leone, M.D. Director of Medical Oncology Rhode Island Hospital Robert M. O'Bryan, M.D. Division Head, Medical Oncology Henry Ford Hospital Jeffrey C. Theiss, Ph.D. Associate Professor and Convenor of Environmental Services University of Texas School of Public Health Joseph N. Gallina, Pharm.D. Secretariat Director of Pharmacy Services University of Maryland Medical System/Hospital National Study Commission on Cytotoxic Exposure Rhode Island Hospital Department of Pharmacy Providence, Rhode Island ~~ 02902 EC EE TS) Supported by a grant from | BRISTOL | [ LABORATORIES | 152 Selected Bibliography Handling Cancer Chemotherapy Agents Adamson, R. H. and Sieber, S. M., “Carcinogenic Po- tential of Cancer Chemotherapeutic Agents in Man,” The Cancer Bulletin 29:179 (6) 1977. * American Society of Hospital Pharmacists, ‘Disposal of Unused Anticancer Drugs,” The Signal May, 1979. Ames, B. N,, et al, “Methods for Detecting Carcino- gens and Mutagens with the Salmonella Mammalian-Microsome Mutagenicity Test,” Muta- tion Research 31:347-364 (December) 1975. ** Anderson, M,, et al., “Development and Operation of a Pharmacy-Based Intravenous Cytotoxic Recon- stitution Service.” B. Med. |. 286:32-36 (Jan. 1) 1983. Anderson, R. W., et al., “Risk of Handling Injectable Antineoplastic Agents,” Am. J. Hosp. Pharm. 39:1881-7 (Dec.) 1982. ** Avis, K. E. and Levchuk, |]. W., “Special Considera- tions in the Use of Vertical Laminar-Flow Work- benches.” Am. |. Hosp. Pharm. 41:81-87 (Jan.) 1984 Ballentine, Ron, “Cancerphobia — or Whatever Hap- pened to Red M&M's?” (editorial) Drug Intell. Clini- cal Pharmacy 16:60-61 (Jan.) 1982. ** Bergemann, D. A., "Handling Antineoplastic Agents.” Am. |. IV Therapy and Clin. Nutrition 10:13-17 (Jan.) 1983 ** Bledsoe, L., “Antineoplastic Agents — Management of Health Risks for the LV. Nurse.” NITA 6:332-333, 1983. Brier, Kenneth L., et al., “Effect of Laminar Air Flow and Clean-Room Dress on Contamination Rates of Intravenous Admixtures,”” Am. |. Hosp. Pharm 38:1144-47 (Aug) 1981 Carrano, A. V,, et al., “Sister Chromatid Exchange as an Indicator of Mutagenesis,” Nature 271:551-553 (Feb. 9) 1978. Clayson, D. B., “Principles Underlying Testing for Carcinogenicity,” Cancer Bulletin 29:161-166 (6) 1977 Crudi, C. B,, “A Compounding Dilemma: I've Kept the Drug Sterile But Have I Contaminated Myself?” NITA 3:77-8, 1980. ** D'Arcy, P. F,, “Reactions and Interactions in Han- dling Anticancer Drugs.” Drug Intell. Clinical Phar macy 17:532-538 (July/Aug.) 1983 * Davis, M. R., “Guidelines for Safe Handling of Cytotoxic Drugs in Pharmacy Department and Hos- pital Wards (The Society of Hospital Pharmacists of Australia),” Hosp. Pharm. 16:17-20 (Jan.) 1981. Donner, A. L., “Possible Risk of Working with An- tineoplastic Drugs in Horizontal Laminar Flow Hoods (letter),”” Am. |. Hosp. Pharm. 35:900 (Aug. 1978. Falck, K., et al, “Mutagenicity in Urine of Nurses Handling Cytostatic Drugs (letter),”” Lancet 1:1250-1 (June 9) 1979 Farber, E., “Chemical Carcinogenesis,” New Engl. | Med. 305:1379-89 (Dec. 3) 1981 ** Gibson, |. F., et al, “Mutagenicity of Urine From Nurses Handling Cytotoxic Drugs” (letter), Lancet 1:100-107 (Jan. 14) 1984 * Gousse, G. C., et al, “Biological Safety Cabinets for Chemotherapy Preparation (letter),” Am. |. Hosp Pharm. 38:967 (July) 1981 Gross, Jody, et al., “Possible Hazards of Working with Cytotoxic Agents,” Oncology Nursing Forum 8(4):10-12, 1981. Hamilton, C. W., “Risk to Personnel Admixing Cancer Chemothrapy,” US Pharm. 7:H1-H8 (July) 1982. Harris, C. C., “A Delayed Complication of Cancer Therapy — Cancer,” [NCI 63:275-7, 1979. * Harrison, Bruce R., “Developing Guidelines for Working with Antineoplastic Drugs,” Am. J. Hosp. Pharm. 38:1686-1693 (Nov.) 1981 ** Hennessy, K. and Duval, A. “Chemotherapy Waste Disposal: A Safe and Practical Method.” NITA 5:311-312, 1982. ** Hirst, M., “Occupational Exposure to Cyclophos- phamide.” Lancet 1:186-188 (Jan. 28) 1984 * Hoffman, D. M., “The Handling of Antineoplastic Drugs in a Major Cancer Center,” Hosp. Pharm 15:302-304 (June) 1980. Hoffman, D. M., “Handling Cytotoxic Drugs (let- ter),” Am. |. Hosp. Pharm. 38:1284-86 (Sept.) 1981. Honda, D., Ignoffo, R. J., and Power, L. A., “Safety Consideration in the Preparation of Parenteral An- tineoplastics,” CSH? Voice 8:94-96 (Winter) 1981. Hoover, R. and Fraumeni, J. D., “Drug-Induced Cancer,” Cancer 47:1071-80 (March 1) 1981 (Sup- plement). ** Johnson, B. L. and Gross, |., “Handling Methotrexate — A Safety Problem,” Am. J. Nursing 82:1531 (Oct.) 1982. Kleinberg, M. L. and Quinn, M. |, “Airborne Drug Levels in a Laminar-Flow Hood,” Am. |. Hosp. Pharm. 38:1301-13 (Sept.) 1981. * Knowles, R. S. and Virden, J. E., “Handling of Injecta- ble Antineoplastic Agents,” Brit. Med. |. 281:589-91 (August 30) 1980. Ladik, C. F, et al, “Precautionary Measures in the Preparation of Antineoplastics (letter),” Am. |. Hosp. Pharm. 37:1184-86 (Sept.) 1980 **LeRoy, M. L., et al, “Procedures for Handling Antineoplastic Injections in Comprehensive Cancer Centers.” Am. |. Hosp. Pharm. 40:601-603 (April) 1983. Marquardt, H. and Marquardt, H. “Induction of Malignant Transformation and Mutagenesis in Cell Cultures by Cancer Chemotherapy Agents,” Cancer 40:1930-34 (Oct.) 1977 (Supplement) ** Mattia, M. A. and Blake S. L., “Hospital Hazards: Cancer Drugs,” Am. |. Nursing 83:758-762 (May) 1983. ** Neal, A. et al, “Exposure of Hospital Workers to Airborne Antineoplastic Agents,” Am. |. Hosp Pharm. 40: 597-601 (April) 1983 ** Nevasaari, K., “Handling Cytostatic Drugs” (letter) Lancet 1:247-248 (Jan. 29) 1983 Ng, L. M., “Possible Hazards of Handling Antineop- lastic Drugs (letter),”” Pediatrics 46:648-49, 1970. Norppa, H., et al, “Increased Sister Chromatid Ex- change Frequencies in Lymphocytes of Nurses Han- dling Cytostatic Drugs,” Scand. |. Work Environ Health 6(4):299-301 (Dec.) 1980. Perlman, M. and Walker, R., “Acute Leukemia Fol- lowing Cytotoxic Chemotherapy (letter),” JAMA 224:250 (April 9) 1973. Redfearn, |J., ‘Safety’ Cabinets are not safe, Says UK Study,” Nature 278:384 (March 29) 1979. Reich, Steven D., “Antineoplastic Agents as Poten- tial Carcinogens: Are Nurses and Pharmacists at Risk?,” Cancer Nursing 4:500-502 (Dec.) 1981. Reimer, R. R. et al, “Acute Leukemia After Alkylat- ing Agent Therapy of Ovarian Cancer,” N. Engl. | Med. 297:177-81, 1977 *References containing handling guidelines ** Additional references since March 1983 Appendix XIII (continued) Rosner, F., “Acute Leukemia as a Delayed Conse- quence of Cancer Chemotherapy,” Cancer 37:1033-6, 1976. Sieber, S. M., “Cancer Chemotherapeutic Agents and Carcinogenesis,” Cancer Chemo. Reports. 59:915-18 (Sept./Oct.) 1975. ** Solimando, D. A. “Preparation of Antineoplastic Drugs: A Review.” Am./. IV Therapy and Clin. Nutri tion 10:16-27 (Sept.) 1983. ** Sotaniemi, E. A., “Liver Damage in Nurses Handling Cytotoxic Agents,” Acta. Med. Scand. 214:181-189, 1983. Staiano, N,, et al, “Lack of Mutagenic Activity in Urine from Hospital Pharmacists Admixing An- titumour Drugs,” Lancet 1:615-16 1981. Stolar, M. H,, et al, “Recommendations for Handling Cytotoxic Drugs in Hospitals,” Am. |. Hosp. Pharm 40:1163-1171 (July) 1983 Stuart, D. G., et al, “Survey, Use, and Performance of Biological Safety Cabinets,” Am. Ind. Hygiene Assoc J. 43:264-670 (April) 1982. Theiss, J. C., “Detection of Carcinogens,”’ in Cancer-Achievements, Challenges, and Prospects for the 1980's, Vol. 1, Burchenal, J. H. and Oettgen, H. F., Editors, Grune and Stratton, New York, 1981, pp 281-90. Thomsen, K. and Mikkelsen, H. I., “Protective Capacity of Gloves Used for Handling of Nitrogen Mustard,” Contact Dermatitis 1:268-69 (Aug.) 1975. * Tortorici, M. P., “Precautions Followed by Personnel Involved with the Preparation of Parenteral Antineo- plastic Medications,” Hosp. Pharm. 15:293-301 June) 1980. ”" ** Vaccari, P. L., et al, “Disposal of Antineoplastic Wastes at the National Institutes of Health,” Am. | Hosp. Pharm. 41:87-93 (Jan.) 1984. ** Venitt, S., et al, “Monitoring Exposure of Nursing and Pharmacy Personnel to Cytotoxic Drugs: Urinary Mutation Assays and Urinary Platinum as Markers of Absorption,” Lancet 1:74-77 (Jan. 19) 1984 Waksvik, H., et al, “Chromosome Analyses of Nurses Handling Cytostatic Agents,” Cancer Treat Rep. 65:607-10 (July/Aug.) 1981. Weisburger, J. H., etal., “The Carcinogenic Properties of Some of the Principal Drugs Used in Clinical Cancer Chemotherapy,” Recent Results Cancer Res. 52:1-17, 1975. Wilson, |. P. and Solimando, D. A., “Antineoplastics A Safety Hazard? (letter),”” Am. |. Hosp. Pharm 38:624 (May) 1981 Wilson, James P., et al, “Aseptic Technique as a Safety Precaution in the Preparation of Antineoplas- tic Agents,” Hosp. Pharm. 16:575-81 (Nov.) 1981 Zellmer, William A., “Reducing Occupational Expo- sure to Potential Carcinogens in Hospitals (edito- rial)” Am. |. Hosp. Pharm. 38:1679 (Nov.) 1981 * Zimmerman, Paul F,, et al, “Recommendations for the Safe Handling of Injectable Antineoplastic Drug Products,” Am. |. Hosp. Pharm. 38:1693-95 (Nov.) 1981 ©c opyright 1984 National Study Commission on Cytotoxic Exposure 153 National Cancer Institute Procedure for Management of Investigational Drugs Acquired for Compassionate Treatment of Individual Patients Food and Drug Administration (FDA) regulations and National Cancer Insti- tute (NCI) policy require the following steps to be completed as indicated: 1) 2 ~~ 3) 4) Statement of Investigator (FDA 1573): Since you are not registered as an investigator, it is required that you complete and sign the enclosed FDA 1573 form and return it within 10 working days in order to receive additional drug. We would suggest that you read this form very carefully since it outlines your responsibilities as a physician receiving investigational drugs. Protocol: A brief protocol must be submitted for each patient which describes the treatment plan, toxicity, efficacy, and monitoring procedures. For your convenience we have devised a standard protocol form which is included and must be completed. The original and three copies must be returned within 10 working days. Please return this document to the following address: Drug Management and Authoriza- tion Section, IDB, CTEP Division of Cancer Treatment, NCI Landow Building, Room 1304-A 7910 Woodmont Avenue Bethesda, MD 20892 Institutional Review Board Approval: You must obtain Institu- tional Review Board approval prior to treatment of the patient and retain documentation of this approval in the patient’s medical record. Informed Consent: You must write an informed consent form which must be signed by the patient and retained in the patient’s medical record. The consent should include a reasonable statement about the side effects of the drug. The informed consent must address each of the eight elements required under FDA regulations. 5) 6) 7) 8) 9) Adverse Drug Reactions: Unexpected toxicity must be reported directly to the NCI. Lethal or life-threatening toxicity must be telephoned to the NCI within 24 hours of its occurrence (301-496-7957, available 24 hours/day). All other toxicities must be reported in writing within 10 days to the Investigational Drug Branch, CTEP, DCT, National Cancer Institute, Landow Bldg., Rm. 4C09, 7910 Woodmont Avenue, Bethesda, MD, 20892. Please read the guidelines for reporting and use the ‘‘Adverse Reaction Form for Investigational Agents’ when reporting adverse drug reactions. Final Patient Report: Upon completion of therapy, you must provide NCI a report of the treatment experience, which describes toxicity and efficacy. We have enclosed the form to be used. Please return this form to the above address. Investigational Drug Accountability: The enclosed investigational drug accountability record must be maintained and retained in your records. These records may be inspected upon request by an authorized representative of the FDA or the NCI. Failure to submit items 1, 2 and 5 in a timely manner will prevent any further drug shipments, and may result in recall of drug. Drug Reorders: Additional drug may be requested only for the same patient previously treated. The patient’s first name and ini- tials of last name should be indicated on the Clinical Drug Request. Telephone orders will not be accepted. December 1985 157 Protocol Amendment No. DEPARTMENT OF HEALTH & HUMAN SERVICES INSTRUCTIONS: 1. Use only this form for Special Exception Protocol. Public Health Service National Institutes of Health 2. Please type. National Cancer Institute SPECIAL EXCEPTION PROTOCOL FOR THE USE OF Investigational Drug(s) NCI Protocol Number Physician’s Name Address Investigator No. PATIENT INFORMATION: Identification (Initials or Identification Number) Age Sex Disease Date of Diagnosis Prior Therapy: JUSTIFICATION FOR SELECTING PLAN OF TREATMENT: 158 Page 2 Special Exception Protocol for use of PLANNED DOSAGE REGIMEN: (Include all chemotherapeutic agents and how often course will be repeated.) DOSAGE MODIFICATION: EXPECTED TOXICITIES: MONITORING CRITERIA: RESPONSIBLE PHYSICIAN Date (Signature) My signature indicates that: 1. Informed consent will be obtained. 2. This protocol will be followed. 3. A report on the patient’s progress will be submitted to the Investigational Drug Branch of the National Cancer Institute after completion of therapy. 4. Any adverse drug reaction encountered will be reported to the Investigational Drug Branch of the National Cancer Institute immediately. 159 Subject Index Adverse Drug Reactions (ADRs) Commercial Drugs and, 11.4 Group C Drugs and, 17.6 Phase I Trials and, 11.1 Phase 11/111 Trials and, 11.2 Protocol Chairman and, 122 Reporting of, 10.2, 10.3. 11, Appendix XI Where to Report 11.3 Affiliate Program Affiliate Investigator Responsibilities 13.3 Drug Ordering by, 9.4 Phase II Trials and, 5.2, Appendix IV Research Base and, 32, 134 Amendments (See Protocol and Information Office) Cancer Therapy Evaluation Program (CTEP) CTEP Letter 45,54,55.,56 as Investigator Drug Sponsor 2 and Phase I Trials 4.1 and Phase II Trials 5.1 and Phase III Trials 6.3 and Protocol Review and Approval 8 Responsibilities for Clinical Trials 2 Scientific Policies of, 4.1, 5.1, 6.1 Clinical Trials Monitoring Service (CTMS) Drug Accountability and Storage 15 Phase I Results Reporting and, 10.2 Phase I and II/III Contractors and, 16.2 Protocol Amendments and, 8.6 Quality Assurance and Monitoring 16, Table 1 Site Visits by, 15,16 Community Clinical Oncology Program (CCOPs) Drug Orders from, 9.3 CTEP Letter and Letters of Intent (LOIs) 5.6 and Phase I Trials 4.5 and Phase II Trials 54,55 Cancer Centers Affiliate Members and, 13.4 Clinical Drug Trials by, 2 Phase II Trials by, 52 as Research Base 3.1 Case Report Forms (See Results Reporting) Centralized Data Management Research Base in 32 Clinical Brochures Phase I Drugs in, 4.5 Phase II Drugs in, 5.5 Clinical Investigations Branch (CIB) Phase III Planning 6.3, Appendix III Clinical Drug Trials Cancer Centers in, 2 Cooperative Groups in, 2, Appendix I Investigator Responsibilities 12.1, 14, Appendix IV New Drug Development Contractors in, 2 Non-Registered Physicians in, 7.2, 14.2 Phase | 4 Phase II 5 Phase 111 6 Pharmaceutical Industry Support of, 24,25, Appendix | Protocol Chairman Responsibilities 12.2 Quality Assurance and monitoring in 16 Sponsors of, 2 University Hospitals in, 2 Compassionate Use of Investigational Drugs (see Group C Drugs, Special Exception Drugs) Cooperative Groups Affiliate Investigators and, 13.4 Clinical Drug Trials by, 2 Letters of Intent (LOIs) from, 5.6 and Pharmaceutical Industry 2.3, Appendix I Phase II Trials by, 52 Phase III Trials by, 6.2 Pharmaceutical Industry and, 2.4 Protocol Compliance and, 16.2 as Research Base 3.1 Drug Ordering Biologic Agents 9.3 Clinical Drug Request Form 9.1, Appendix X Group C 9.2, 95, 17.5 for Non-Clinical Use 9.7 Procedures for, 9,93 Special Exception 9.2, 95, 18.3 Drug Accountability and Storage Drug Returns 15.2, Appendix X NCI Drug Accountability Record Form 15, Appendix XII Procedures for, 15 Research Base in, 3.2 Safe Handling 15.4, Appendix XIII Site Visits and, 15.3, 16.4 Drug Distribution and Protocol Face Sheet 71 to Registered Investigators 14.1, Appendix IV Drug Accountability Record Form (DARF) Accountability and Storage 15, Appendix XII and Group C Drugs 17.6 Funding Clinical Trials National Cancer Advisory Board (NCAB) and, 21 Peer Review and, 2.1 Food and Drug Administration (FDA) INDs and, 2.6 NDAs and, 27 Oncologic Drugs Advisory Committee of, 2.7 Regulations and NCI 2 Regulations and Pharmaceutical Industry 2 Regulations and Clinical Investigators 2 Results Reporting Requirements of, 10, 10.4 Statement of Investigator (FDA-1573) 12.1, 13.3, 14.1, 17.5, Appendix IV 161 162 FDA-1573 (Statement of Investigator, see above) Freedom of Information Act and Patient Medical Records 16.6 Group C Drugs Guideline Protocols for, 17.4 Ordering of 9.2, 17.5 Responsibilities for use, 17.6 Informed Consent Document and Affiliate Investigators 13.3 Checklist Appendix VII and Group C Drugs 17.6 and Investigator Responsibilities 121 and Monitoring Program 16.4, 16.6 and Protocol Review Committee (PRC) 8.4 and Protocol Specificity 7.3 Institutional Review Board (IRB) and Affiliate Investigators 13.3 and Group C Drugs 17.6 in Monitoring and Quality Assurance 16.4 Protocol Chairman and, 12.2 and Protocol Review and Approval Process 32,33 and Results Reporting 10.5 Investigational Drug Administration by Non-Registered Physicians 7.2, 14.2 by Registered Physicians 14.1, Appendix IV Investigational Drug Branch (IDB) as Drug Information Source 45, 5.5, Appendix III and Drug Ordering 9.1, Appendix III and Letters of Intent (LOIs) 5.6, Appendix III and Phase III Planning 6.3, Appendix III Investigational New Drug (IND) Applications FDA and, 2,26 Sponsor Responsibilities for, 2.6 Investigator Responsibilities in Clinical Trials 12.1, Appendix IV for Affiliate Investigators 13.4 Drug Accountability and Storage 15, Appendix XII Research Base and, 3.1 Results Reporting to CTEP 10 Results Reporting to IRBs 10.5 Letters of Intent (LOIs) 5.6, Appendix VI Monitoring and Quality Assurance Informed Consent Document and, 16.6 Multicenter Trials, and, 72 Research Base in, 3.2, 16 Site Visit Auditing Program 16.5 Multicenter Trials Phase 11 52, 5.3 Phase 111 6.2 and Protocol Development 72 New Drug Application (NDA) Commercial Drug Availability and, 27 FDA and, 2,27 Private Sponsor Responsibility for, 27 New Drug Study Groups and Phase II Trials 5.2, Appendix V Office of Protection from Research Risks (OPRR) Affiliate Investigators and, 13.2 Human Subjects and, 2 Investigator Responsibilities and, 12.1 Pharmaceutical Industry Cooperative Groups and, 24 Federally Funded Trials and, 24 Federally Sponsored Unfunded Trials and, 2.5 NDA Submissions and, 27 Phase I Contractors and, 24 Phase 11/111 Contractors and, 24 Phase I Trials Ad Hoc Investigators and, 42 Children in, 4.3, Appendix II Contract Programs for, 4.2 CTEP Scientific Policies for, 4.1 Drug Information for, 4.5, Appendix III Institutional Eligibility for, 43 Planning 4.1 Reporting Adverse Drug Reactiosn (ADRs) 11.1 Results Reporting 10.2 Phase II Trials Cancer Centers and, 5.2 Cooperative Groups and, 5.2 CTEP Scientific Policies for, 54 Drug Combinations in, 5] Institutional Eligibility for, 52 Letters of Intent (LOIs) in, 5.6 Multicenter Conduct of, 52 Phase 11/111 Contractors for, 52 Planning 5.1 Prior Therapy and Patient Eligibility 5.1 Reporting Adverse Drug Reactions (ADRs) 11.2 Results Reporting 10.3 Single Drugs in, 5.1 Phase III Trials Control Groups in, 6.1 Cooperative Groups and, 6.2 CTEP Scientific Policies for, 6.1 Multicenter Approach to, 6.2 Planning 6.3 Reporting Adverse Drug Reactions (ADRs) 11.2 Results Reporting 10.3 Preclinical Drug Development Biological Response Modifiers Program and, 2.2 DCT and, 22 Protocol Chairman Clinical Trials Responsibilities 12.2 Protocol Data Query (PDQ) and Protocol Face Sheet 7.1 and Protocol and Information Office Protocol Development Consensus Review 8.4 Drafting of Protocols 7 Essential Protocol Elements 7.2 Informed Consent and, 7.3, Appendix VII and Multicenter Trials 7.2 and Phase I Trials 4.1 and Phase II Trials 5.1 Protocol Submission Checklist 8.1, 8.2, Appendix VIII Protocol Face Sheet Zl Research Base and, 3.2 Review and Approval Process 8 Statistical Considerations in, 51,61, 72 Protocol and Information Office and Letters of Intent (LOI) 5.6 Appendix VI and Protocol Amendments 8.6, 10.3 and Protocol Face Sheet 7.1 and Protocol Review and Approval 8 and Protocol Study Status 8.7, 8.8, 10.3 Appendix VIII and Results Reporting 10.3.2 and Publications 10.2.7 Protocol Review Committee and Letters of Intent (LOIs) 5.6 in Protocol Review and Approval 8.4 Records Retention FDA Requirements for, 10.4 Referring Physicians and Investigational Drug Administration 7.2, 14.1 Research Base Clinical Trials Support by, 32 Definition and Responsibilities 3.1 Drug Ordering and, 9.1 Monitoring and Quality Assurance by, 16 Responsibility for Affiliates 13.4 Results Reporting by, 32 Results Reporting Case Report Forms 10.1, 10.2, 10.3 Phase I Trials 10.2 Phase 11/111 Trials 10.3 Publications 10.2, 10.3 Research Base and, 3.2 Study Summary Forms 10.1, 10.2, 10.3 Site Visits Drug Accountability and Storage 15 Quality Assurance and Monitoring 16.5, Table 1 Special Exception Drugs Ordering 9.2, 18.3 Responsibilities of use Appendix XIV Sponsor of Drug Trials Collaborative Efforts 2.3, Appendix I Defined 2 Division of Cancer Treatment (DCT) as, 23 IND Submission by 2.6 Monitoring and Quality Assurance 16 Pharmaceutical Firms as, 23 Study Summary Forms (see Results Reporting) 163 U.C. BERKELEY LIBRARIES TA 8001138918