throbber
Cancer Chemother Pharmacol (1998) 41: 173±185
`
`Ó Springer-Verlag 1998
`
`O R I G I N A L A R T I C L E
`
`Joseph J. DeGeorge á Chang-Ho Ahn
`Paul A. Andrews á Margaret E. Brower
`Diana W. Giorgio á M. Anwar Goheer
`Doo Y. Lee-Ham á W. David McGuinn
`Wendelyn Schmidt á C. Joseph Sun
`Satish C. Tripathi
`Regulatory considerations for preclinical development
`of anticancer drugs
`
`Received: 19 December 1996 / Accepted: 2 June 1997
`
`Abstract The entry of new anticancer treatments into
`phase I clinical trials is ordinarily based on relatively
`modest preclinical data. This report de®nes the battery
`of preclinical tests important for assessing safety under
`an Investigational New Drug application (IND) and
`outlines a basis for extrapolating starting doses of in-
`vestigational anticancer drugs in phase I clinical trials
`from animal toxicity studies. Types of preclinical studies
`for the support of marketing of a new anticancer drug
`are also discussed. This report addresses di€erences and
`similarities in the preclinical development of cytotoxic
`drugs (including photosensitizers and targeted delivery
`products), drugs used chronically (chemopreventive
`drugs, hormonal drugs, immunomodulators), and drugs
`intended to enhance the ecacy (MDR-reversing agents
`and radiation/chemotherapy sensitizers) or diminish the
`toxicity of currently used anticancer therapies. Factors
`to consider in the design of preclinical studies of com-
`bination therapies, alternative therapies, and adjuvant
`therapies in the treatment of cancer, and to support
`changes in clinical formulations or route of adminis-
`tration, are also discussed.
`
`Key words Antineoplastic agents á Toxicity tests á
`Toxicology á Guidelines á Phase I clinical trials
`
`This article is not an ocial FDA guidance or policy statement. No
`ocial support or endorsement by the Food and Drug Adminis-
`tration is intended or should be inferred
`
`J.J. DeGeorge á C-H. Ahn á P.A. Andrews (&)
`M.E. Brower á D.W. Giorgio á M.A. Goheer á
`D.Y. Lee-Ham á W.D. McGuinn á W. Schmidt á
`C.J. Sun á S.C. Tripathi
`Division of Oncology Drug Products,
`Center for Drug Evaluation and Research,
`Food and Drug Administration, 5600 Fishers Lane HFD-150,
`Rockville, Maryland 20857, USA
`Tel. (301)-594-5782; Fax: (301)-594-0499;
`E.mail: andrewsp@cder.fda.gov
`
`Introduction
`
`Malignant, nonresectable cancers are life-threatening,
`and aggressive measures are used in treating them.
`Antineoplastic
`therapies
`frequently
`include
`toxic
`chemicals or biological products that are designed to
`destroy tumor tissue or halt cell replication. Despite
`the serious toxicities of many anticancer drugs, careful
`dosing, clinical monitoring and prompt treatment of
`toxicity makes the side e€ects less threatening to a
`patient than their disease. Since it is recognized that
`doses of anticancer drugs high enough to kill cancer
`cells usually induce serious side e€ects in patients, the
`preclinical testing of oncology drugs di€ers from test-
`ing of nononcology drugs. The Division of Oncology
`Drug Products within the Center for Drug Evaluation
`and Research (CDER) at the US Food and Drug
`Administration (FDA) recognizes the urgency of de-
`velopment of new anticancer drugs and the need to
`rapidly move promising agents into clinical studies.
`This report o€ers a regulatory perspective on the pre-
`clinical development of new anticancer drugs that is
`intended to clarify the di€erences from the preclinical
`testing of nononcology drugs and to describe the data
`that are important to support human testing and even-
`tual marketing.
`The types of preclinical studies expected for support
`of clinical trials and then marketing of a new drug de-
`pend on both the intended use of the drug and the
`population of patients being studied and treated. In
`situations where potential bene®ts are greatest (ad-
`vanced, life-threatening disease), greater risks of treat-
`ment
`toxicity can be accepted and the required
`preclinical testing can be minimal. In cases where the
`patient population is free of known disease (e.g. adju-
`vant therapy or chemoprevention) the acceptable risks
`are much less and preclinical evaluation should be
`more extensive [32]. The toxicities of many modulating
`agents intended to enhance the ecacy or diminish the
`toxicity of anticancer agents are more similar to those of
`Genentech 2133
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`174
`
`these modulating
`nononcology therapies. However,
`agents could enhance the toxicity or diminish the activity
`of cytotoxic drugs by altering their toxicodynamics,
`pharmacodynamics, and pharmacokinetics. Thus, toxi-
`cological evaluation in combination with the modulated
`cytotoxic drug is an important part of preclinical de-
`velopment.
`The following considerations are o€ered in an e€ort
`to balance the risks to be borne by the proposed patient
`population and the realities of drug testing in humans.
`The di€erences in preclinical testing between cytotoxic,
`chronic (i.e. adjuvant therapy, chemopreventive drugs,
`hormonal drugs, and immunomodulators), and modu-
`lating therapies are emphasized. Issues of chemistry and
`manufacturing controls, clinical study design, and de-
`velopment of biologic agents for cancer treatment are
`beyond the scope of this report. If the appropriate
`preclinical development strategy remains uncertain after
`contemplating
`the
`following
`considerations,
`then
`sponsors are encouraged to initiate pre-IND discussions
`with Division sta€ regarding their preclinical study
`plan.
`
`General considerations for anticancer drug development
`
`Preclinical studies of anticancer agents
`
`The safety of ®rst-time use in humans is assessed
`through preclinical
`studies of pharmacodynamics,
`pharmacokinetics (toxicokinetics), toxicity, and their
`relationships. The purposes of these safety studies are:
`(a) to determine a starting dose for clinical trials that is
`both reasonably safe and allows for possible clinical
`bene®t for the patient, (b) to identify potential end-or-
`gan toxicities and determine their reversibility, and (c) to
`assist in the design of human dosing regimens and es-
`calation schemes for clinical
`trials. Animal
`toxicity
`studies most e€ectively accomplish these objectives when
`performed using schedules, durations, formulations, and
`routes comparable to those proposed in clinical studies.
`Use of longer duration preclinical studies may lead to
`underestimates of the appropriate clinical dose, while
`shorter studies may not identify cumulative dosing tox-
`icities. The toxicity studies should generally conform to
`the protocols recommended by the National Cancer
`Institute
`for
`toxicology assessment
`for anticancer
`agents1 and are expected to be conducted in accordance
`with Good Laboratory Practices (GLP) [16, 17]. When
`studies are not performed according to GLP, deviations
`should be documented and the potential impact of these
`deviations on study outcome and credibility should be
`described [16, 17].
`Typically, only two toxicology studies are essential to
`support initial phase I clinical trials in patients with
`
`1 The Developmental Therapeutics Program; Division of Cancer
`Treatment, Diagnosis, and Centers; National Cancer Institute
`(Rockville, MD USA) may be contacted for protocol details
`
`advanced cancers (Table 1). The ®rst of these is usually a
`study in rodents that identi®es doses that produce life-
`threatening and non-life-threatening toxicity. The sec-
`ond study should determine whether doses identi®ed as
`tolerable in rodents produce life-threatening toxicity in a
`non-rodent species. At least one of these studies should
`assess clinical signs, body weight, food consumption,
`clinical pathology, and gross pathology over a range of
`doses from nontoxic to toxic and should include an ex-
`amination of histopathology at doses that cause toxicity
`(or at the highest dose tested). Genotoxicity tests are not
`generally needed for cancer chemotherapies to support
`testing in phase I clinical studies unless healthy volun-
`teers will be entered into the study.
`While not essential, information on the pharmaco-
`dynamics and pharmacokinetics of drugs is extremely
`valuable for supporting the safety pro®le and can sig-
`ni®cantly contribute to the eciency of drug develop-
`ment. A phase I study may be conducted with no in vitro
`or in vivo preclinical pharmacodynamic information,
`but preclinical studies on biological activity and ecacy
`can substantially aid in clinical study design. Such
`studies help estimate e€ective dosages, dosing schedules,
`and optimal plasma concentrations. This information is
`likely to be particularly useful when developing noncy-
`totoxic agents. It may be desirable to develop such
`agents (e.g. immunomodulators) by escalating the hu-
`man dose to a pharmacodynamically active range rather
`than to the maximum tolerated dose (MTD). Pharma-
`cokinetic data can be gathered as a part of pharmacol-
`ogy or toxicity studies and do not usually need to be
`collected separately. Single- and multiple-dose pharma-
`cokinetic studies in the most appropriate species are best
`performed using dosing schedules, durations, and routes
`comparable to those that will be used in clinical studies
`[15]. The pharmacokinetic information obtained assists
`the evaluation of animal toxicity and ecacy, and may
`suggest modi®cations in the intended dose, route or
`schedule for the clinical trial. The importance of the
`parameters being measured will vary depending on the
`clinical trial design and therapeutic classes as discussed
`in the subsections below. In combination with pharma-
`codynamic data, this information can be used to help
`calculate initial doses in humans that have a greater
`likelihood of activity without adversely a€ecting safety,
`and can contribute to optimal dose escalation in early
`clinical studies.
`The proposed therapeutic indication, the outcome of
`early clinical development, the nature of toxicities seen
`in animals and in humans, and the projected duration
`of clinical treatment all determine the preclinical studies
`necessary to support a New Drug Application (NDA).
`In general, for oncology drugs, sponsors should con-
`duct toxicity studies using the same schedule and du-
`ration of administration as
`the
`intended clinical
`treatment cycle (Tables 1±3). Cytotoxic drugs used to
`treat advanced disease rarely need studies with more
`than 28 days of dosing submitted with the NDA (Ta-
`ble 1). In contrast, for drugs intended for continuous
`
`

`

`Table 1 Preclinical studies for cytotoxic oncology drugs
`
`Stage
`
`Category
`
`Issues to be addressed
`
`Studies considered importanta
`
`IND
`
`All cytotoxics
`
`Modi®cations for
`Special Categories
`Photosensitizer
`
`Starting dose,
`end-organ toxicities
`Genetic toxicity
`E€ective concentrations,
`schedule
`
`Systemic toxicity
`
`Phototoxicity
`
`Plasma t1/2
`
`Rodentb and nonrodentc
`toxicologyd
`Genetic toxicity panele
`
`Toxicology studies
`in subdued light
`
`Antibody conjugate
`
`Stability
`Toxicity of drug alone
`Speci®city
`
`Stability in plasma
`Toxicology in one species
`Human tissue screen
`
`Pharmacokinetics
`
`Liposomal delivery
`
`Drug product toxicity
`
`Depots
`
`Pharmacokinetics versus
`free drug
`Drug product toxicity
`
`Include free drug and blank
`liposomes in toxicity testing
`Pharmacokinetics
`
`Include free drug and
`empty depot in toxicity testing
`
`Toxicity to contacted tissues
`
`Histopathology of depot site
`
`175
`
`Studies considered
`useful
`
`Pharmacokinetics,
`pharmacodynamics
`
`In vivo study with
`illuminated skin
`Pharmacokinetics
`
`Activity in cell lines
`‹ target antigen
`
`Pharmacokinetics
`
`NDA All cytotoxics
`
`Rodent and nonrodent
`toxicologya,g,
`genetic toxicity,
`stage C-D teratogenicityf
`in rodents and nonrodents
`
`Targeted special
`toxicity
`
`a In general, the schedule and duration of administration in the toxicology study should mimic the clinical trial
`b Should determine the dose severely toxic to 10% of the animals (STD10)
`c Should determine toxicity of one-tenth the rodent STD10 on a mg/m2 basis
`d One study should include histopathology
`e Only for phase I testing in normal volunteers or patients believed to be disease-free
`f Should be submitted during development
`g Studies with more than 28 days of dosing are rarely needed
`
`daily administration such as for chemoprevention, ad-
`juvant therapy, or long-term hormonal or immuno-
`modulation
`therapy,
`chronic
`studies
`should
`be
`conducted up to a maximum of 6 months in rodent and
`12 months in nonrodent species (Table 2). International
`Conference on Harmonization (ICH) stage C-D2 re-
`productive toxicity studies in a rodent and a non-rodent
`species are important components of the preclinical
`evaluation of anticancer drugs and should be submitted
`early in development [14].
`Carcinogenicity studies are not required for cytotoxic
`drugs used to treat advanced systemic disease, but can
`
`2 ICH stage A-B, C-D, and C-F reproduction toxicity studies
`correspond to the previously designated segment I, II, and III
`studies which are de®ned by daily administration of drug,
`respectively, during the period from premating to implantation,
`implantation to birth (period of organogenesis), and implantation
`to sexual maturity [14]
`
`be important in the assessment of drugs intended for
`chronic use for chemoprevention, adjuvant, or hor-
`monal therapy when patients are likely to have a long
`survival [18]. The current standard is the 2-year rodent
`bioassay [47], although alternatives may be suitable [20].
`Depending upon the nature of toxicities seen with the
`drug or drug class in animals and in humans, targeted
`special toxicity studies to support NDA ®ling may also
`be needed. For example, in the development of anthra-
`cyclines and platinum drugs, which are known to have
`cardiotoxic and ototoxic potential, respectively, addi-
`tional preclinical cardiotoxicity and ototoxicity studies
`have been useful [11, 28, 36, 46]. In addition, neonatal
`reproductive toxicology and DNA adducting studies
`have been useful in the development of antiestrogenic
`agents [5, 30, 31, 37, 44, 45]. A discussion with FDA sta€
`on the preclinical studies needed for marketing approval
`for a particular drug is recommended at or before the
`end of phase II clinical studies.
`
`

`

`176
`
`Table 2 Preclinical studies for noncytotoxic, chronically administered oncology drugs
`
`Category
`
`Studies considered important
`
`Studies considered useful
`
`Stage
`
`IND
`
`All noncytotoxic chronic
`therapy
`
`Modi®cations for special
`categories
`Adjuvant therapy
`
`Chemopreventive
`
`Hormonal
`
`Immunomodulator
`
`Rodenta and nonrodentb
`toxicologyc,d
`
`Pharmacokinetics,
`-dynamics
`
`Ecacy studies
`
`Carcinogenicitye
`Stage A-B reproductive toxicity
`Stage C-D teratogenicitye
`
`Genetic toxicity panele
`
`Toxicology studies should
`also de®ne NOAEL,
`Genetic toxicity panel
`
`28-day toxicology studies usually suce for
`limited phase I/II testing in advanced cancer,
`genetic toxicity panele
`
`28-day toxicology studies usually suce for
`limited phase I/II testing in advanced cancer,
`genetic toxicity panele, de®ne dose versus
`immunologic response curve to identify shape
`(bell-shaped?) and surrogate markers
`
`Toxicology studies of equivalent duration to
`labeled use up to 6 months in rodents and
`12 months in nonrodents, genetic toxicity panel,
`carcinogenicityf, stage C-D teratogenicity in
`rodents and non-rodents
`
`NDA
`
`All non-cytotoxic chronic
`therapy
`
`Additional for hormonal
`
`Stage A-B reproductive toxicity
`
`Stage C-F reproductive toxicity,
`neonatal reproductive
`tract toxicity, DNA
`adducting (drug speci®c)
`
`Additional for
`chemopreventive
`
`Stage A-B and C-F reproductive toxicity
`carcinogenicity (always)
`
`a Should determine the dose severely toxic to 10% of the animals (STD10)
`b Should determine toxicity of one-tenth the rodent STD10 on a mg/m2 basis
`c In general, the schedule of administration in the toxicology study should mimic the clinical trial with a duration as long as the intended
`clinical study up to 6 months in rodents and 12 months in non-rodents
`d One study should include histopathology
`e Expected prior to clinical testing in patients with low risk of cancer recurrence, or testing in healthy volunteers
`f May be unnecessary depending on intended patient population [18]
`
`Starting doses and dose escalation
`
`As described above, one of the primary goals of preclin-
`ical studies is to estimate a safe starting dose for the ini-
`tiation of phase I trials in humans. The starting dose for
`clinical trials with cytotoxic drugs for oncology indica-
`tions has traditionally been one-tenth the dose lethal to
`10% of rodents on a body surface area basis (milligrams
`per meter squared) [23, 29, 35]. Studies that actually
`measure death as an endpoint, however, are not required
`so long as the dose range studied includes doses that cause
`severe, life-threatening toxicity. Thus, the starting dose is
`generally now chosen as one-tenth of the dose that causes
`severe toxicity (or death) in 10% of the rodents (STD10)
`
`3 This calculation is the same as taking one-third of the toxic dose
`low (TDL) [29, 35]. We believe the current expression of ``one-sixth
`the highest non-severely toxic dose'' is simpler and can be applied
`to the data more universally than taking, in practice, ``one-third the
`dose which causes toxicity but when doubled does not kill the non-
`rodents''. Frequently, the TDL cannot be technically de®ned in
`many studies
`
`on a milligrams per meter squared basis, provided that
`this starting dose, i.e. one-tenth the STD10, does not cause
`serious irreversible toxicity in a nonrodent species [29,
`35]. If irreversible toxicities are produced at the proposed
`starting dose in nonrodents (usually dogs) or if the non-
`rodent is known to be the more appropriate animal
`model, then the starting dose would generally be one-
`sixth of the highest dose tested in nonrodents that does
`not cause severe, irreversible toxicity3. In some cases,
`rodents or dogs may not be appropriate species because
`they do not model the relevant human biochemical or
`metabolic processes. For example, folate pools in rodents
`greatly exceed those in humans [4], so that rodents are
`generally inappropriate species for testing antifolates.
`Also, dogs poorly predict the toxicity of some platinum
`analogues, and an alternate animal model might be pre-
`ferred [34]. Knowledge of relevant physiological, bio-
`chemical, and pharmacokinetic di€erences between
`humans and animal models can help determine the most
`appropriate species to be used for selecting a starting
`dose. Whenever feasible, these starting doses should be
`
`

`

`Table 3 Preclinical studies for modulators of oncology drugs
`
`Stage
`
`Category
`
`Issues to be addressed
`
`Studies considered importanta
`
`IND
`
`All modulators
`
`Starting dose,
`end-organ toxicities
`
`Genetic toxicity
`E€ective concentrations,
`schedule
`
`Rodentb and
`non-rodentc
`toxicologyd
`genetic toxicity panele
`
`Additional studies for
`special categories
`MDR modulator
`
`Combination toxicity
`
`Chemosensitizer
`
`Combination toxicity
`
`Pharmacokinetic perturbations
`
`One species at minimally
`and signi®cantly toxic
`doses of cytotoxic
`Pharmacokinetics
`
`One species at minimally and
`signi®cantly toxic doses of
`cytotoxic
`
`Radiation sensitizer
`
`Delayed toxicity to normal
`tissues
`
`Chemoprotection
`
`Combination toxicity, tumor
`protection
`
`In vivo ecacy of combination
`with histopathology
`
`NDA All modulators
`
`Toxicology studies of equivalent
`duration to labeled use up to
`6 months in rodents and 12 months
`in non-rodents, genetic toxicity,
`stage C-D teratogenicity in rodents
`and non-rodents
`
`a In general, the schedule and duration of administration in the toxicology study should mimic the clinical trial
`b Should determine the dose severely toxic to 10% of the animals (STD10)
`c Should determine toxicity of one-tenth the rodent STD10 on a mg/m2 basis
`d One study should include histopathology
`e Only for phase I testing in normal volunteers or patients believed to be disease-free
`
`177
`
`Studies considered
`useful
`
`Pharmacokinetics
`
`In vivo ecacy of
`combination
`
`Skin/leg contracture
`
`Targeted special
`studies
`
`calculated from studies using the proposed clinical route,
`schedule, and duration.
`The dose escalation scheme for phase I clinical studies
`often follows the standard or modi®ed Fibonacci proce-
`dure [10]. Examples of other common and acceptable
`approaches include modi®ed continual reassessment
`methods [13, 39] and pharmacokinetically guided dose
`escalation strategies [8]. These alternatives often necessi-
`tate a more extensive preclinical evaluation. For example,
`pharmacokinetic guidance of dose escalation is most ef-
`fectively applied when: (a) linear pharmacokinetics are
`observed at drug concentrations spanning the pharma-
`cological and toxicological e€ects, (b) the area under the
`drug concentration versus time curve (AUC) at the mouse
`STD10 can be de®ned, (c) protein binding in mouse and
`human plasma has been quanti®ed, and (d) it is known
`whether metabolites contribute to the toxic e€ects [7, 8,
`27, 40]. Although preclinical studies are used to determine
`the starting dose for phase I clinical trials, the highest
`doses for oncology drugs are rarely restricted by the doses
`used in preclinical toxicology studies as long as the tox-
`icities of the new anticancer drug can be readily moni-
`tored, are reversible, and suciently precede lethality in
`animals. Instead, the maximum dose is restricted by the
`toxicity observed in the clinical trial, judged most often
`using NCI/DCTDC Common Toxicity Criteria [38].
`
`Considerations for speci®c cytotoxic therapies
`
`Combinations of cytotoxic agents
`
`The evaluation of cytotoxic agent combinations has
`traditionally been conducted in the clinical setting using
`an empirical approach. This has generally been suc-
`cessful, but may not be optimal. Preclinical studies
`provide an opportunity to explore a variety of doses,
`dose ratios, and schedules to optimize bene®t and min-
`imize toxicity. Nonetheless, unless there is reason to
`believe that synergistic interactions occur that would
`substantially increase the toxicity of the combination,
`preclinical testing is not considered essential provided
`that each agent has been fully evaluated in humans.
`When synergistic e€ects may be anticipated such as
`when one agent interferes with the metabolism or elim-
`ination of the other agent or both cytotoxic agents target
`the same metabolic pathway or cellular function, pre-
`clinical testing of the combination is desirable.
`
`Photosensitizers
`
`One class of cancer chemotherapeutic drugs is thera-
`peutically inactive until
`irradiated with light. These
`
`

`

`178
`
`photosensitizers or phototherapy agents usually form
`radicals after absorbing light energy that are ultimately
`responsible for tumor destruction. In photosensitizer
`therapy, tumor tissues are typically irradiated with laser
`light. When there is a choice, longer wavelengths of the
`irradiating light are preferred because they cause less
`direct tissue damage and because they penetrate more
`deeply into tumor tissue than shorter wavelengths.
`Selective damage to tumor tissue is obtained by di-
`recting the activating light to the tumor. In addition,
`most phototherapy compounds concentrate in tumor
`tissues more than in surrounding normal tissue when
`given systemically. This increased concentration of
`photosensitizer combined with localized irradiation can
`kill tumor cells with great selectivity. Nevertheless, when
`these compounds are given systemically they commonly
`distribute in appreciable concentrations in all tissues and
`this provides the potential for toxicity. When these drugs
`accumulate in the eye or skin, patients may su€er irre-
`versible retinal damage or severe phototoxicity similar to
`sunburn when exposed to ambient light [12]. Thus, it is
`important to know the plasma elimination half-life (and,
`if possible, tissue elimination half-lives) in preclinical
`studies so that the length of time a patient should protect
`themselves from light can be estimated.
`Standard toxicity studies with multiple dose levels
`should be conducted in subdued illumination to clearly
`de®ne the systemic toxicities of the photosensitizer. Sub-
`dued lighting allows systemic toxicities to be more clearly
`distinguished from phototoxicities. In addition to these
`standard toxicity studies, it is bene®cial to assess photo-
`toxicity before phase I clinical investigation begins be-
`cause these drugs can cause prolonged photosensitivity.
`Acceptable models for these photosensitivity tests are
`either hairless or appropriately shaved species. The pho-
`tosensitivity assessment should include toxicity testing as
`a function of both light dose (total energy) and drug dose
`and should ideally determine the duration of sensitivity in
`relation to plasma levels of the photosensitizer. Since a
`primary concern for the patient is the toxicity related to
`sunlight exposure, the light source for these tests should
`have a spectral distribution that approximates sunlight.
`Frequently, doses that are well below the no observable
`adverse e€ect limit (NOAEL) when the animal is housed
`in subdued light are lethal when the animal is brie¯y ir-
`radiated. Even though the photodynamic e€ect is ex-
`pected to a€ect only tissues that are exposed to the light
`source, there is concern that photodegradation products
`could cause distant toxicities. Therefore, these photo-
`toxicity tests usually include standard assessments of
`clinical signs, clinical pathology, gross pathology, histo-
`pathology of major organs, and the reversibility of tox-
`icities. Clinical photodynamic therapy does not routinely
`involve repeated doses, and thus preclinical studies using
`daily irradiation during repeat dose testing may not be
`relevant to clinical safety concerns.
`Without light these photosensitizers may not cause
`genotoxicity in standard tests, but subsequent irradia-
`tion may cause considerable damage to the DNA of cells
`
`exposed to the compound. Thus, genotoxicity tests are
`best done with and without light. The assessment of
`clastogenicity and mutagenicity should be done with
`increasing compound concentrations at a high light
`dose, and with increasing light dose (total energy) using
`broad-spectrum light at high compound concentration.
`The highest doses of drug of each series of tests should
`be consistent with international standards [19, 22].
`In many cases an e€ective dose of drugs in this class is
`nontoxic in subdued light and the starting dose can be
`chosen based on ecacy studies rather than toxicity
`studies. This pertains only if the projected ecacious
`starting dose is lower than the safe dose estimated from
`the toxicity studies.
`
`Specialized drug delivery
`
`Administration of anticancer drugs as depots, attached
`to carriers, or in specialized encapsulated forms has the
`potential for signi®cantly improving ecacy. Advan-
`tages of specialized drug delivery may include: (a) spe-
`ci®c targeting of the drug to the tumor, (b) minimization
`of toxic side e€ects, (c) prolongation of therapeutic drug
`concentrations, (d) improved delivery of hydrophilic
`drugs to tumor cytoplasm, and (e) practical adminis-
`tration of very lipophilic drugs. Examples of delivery
`systems include copolymer implants, human albumin
`microspheres, monoclonal antibody±drug conjugates,
`and liposomal encapsulation. Development of antican-
`cer drugs administered via carriers or in depots may
`necessitate additional preclinical evaluation beyond that
`of conventional cytotoxic drugs.
`For antibody±drug conjugates, the two main safety
`concerns are the potential for toxicity from abrupt re-
`lease of the drug and the potential for the antibody±drug
`conjugate to cause unexpected, speci®c toxicity in nor-
`mal human tissues. Studies of the stability of the con-
`jugate in human plasma as a function of the proposed
`release mechanism (e.g. pH if hydrolytic, glutathione
`concentration if reductive) help determine the necessity
`of conducting additional toxicology studies [21]. When
`additional studies are indicated, using the form of the
`drug released from the conjugate (i.e. including linker
`groups) may identify clinically important
`toxicities.
`Testing the reactivity of the conjugate with a complete
`panel of human tissues from at least three di€erent
`sources is suggested [21]. When the target antigen is not
`expressed in the tissues of the standard preclinical ani-
`mal models, a tolerance study in Pongidae apes at a dose
`that is at least double the planned human starting dose
`should also be considered. Both the reactivity screen
`and the tolerance study may reveal sites of potential
`tissue-speci®c toxicity, while the standard toxicology
`studies may de®ne nonspeci®c toxicities. Speci®city
`studies of binding or cytotoxicity in cell lines with and
`without an expressed target antigen also help to assess
`whether there is a signi®cant di€erential between the
`toxicity to a targeted and nontargeted tissue. If feasible,
`
`

`

`pharmacokinetic studies that distinguish between con-
`jugate, free antibody, and free drug are also highly
`desirable for interpreting toxicology ®ndings and sup-
`porting interspecies comparisons. Selection of a starting
`dose for clinical study should consider not only the
`results of the toxicity studies with the conjugate, but
`also the stability of the conjugate and the potential
`toxicity of released drug.
`With liposomal drugs, standard preclinical toxicol-
`ogy studies of the delivery system, free drug, and the
`®nal formulation are important for evaluating a drug
`product's potential for toxicity. Liposomal formula-
`tions usually dramatically prolong systemic exposure.
`Thus, when repeated doses are to be used clinically, it is
`especially important to study a similar schedule pre-
`clinically because of the potential for drug accumula-
`tion. When the delivery system is designed to a€ect
`drug absorption, distribution, biotransformation, ex-
`cretion or target organ accumulation, small changes in
`the design of the delivery system may have substantial
`e€ects on overall
`toxicity. Conducting the toxicity
`studies with the ®nal formulation can avoid concerns
`about
`such e€ects. Comparative pharmacokinetic
`studies of the ®nal formulation versus free drug can be
`very helpful
`in suggesting schedules and interpreting
`changes in the spectrum and severity of toxicities. Oc-
`casionally, studies of the empty liposomes plus free
`drug in combination may also be useful for under-
`standing alterations in ecacy seen with the liposomal
`preparation. For example, blank liposomes may alter
`the pharmacokinetics of the free drug in a fashion
`sucient for therapeutic gain [33].
`Preclinical development of depot formulations gen-
`erally follows that of liposomal formulations. Addi-
`tionally, a study of the toxicity of the depot in the tissue
`or compartment intended to be used clinically should be
`conducted which includes a histopathologic examination
`of the adjacent tissues. Initial clinical doses similar to the
`total dose of the drug previously investigated in humans
`may be used in the absence of signi®cant changes in
`toxicity pro®le for the depot formulation.
`
`Alternative therapies
`
```Alternative'' therapies include both single agents and
`multicomponent entities derived from plants or animals.
`Herbal products and tissue or ¯uid extracts from animal
`sources intended for the treatment or prevention of
`cancer or precancerous conditions belong in this cate-
`gory. The identity of the active ingredient of these en-
`tities is frequently uncertain. Consistency in taxonomic
`identi®cation, collection, storage, and processing may
`pose additional diculties. A useful initial step is to
`prepare a batch of the drug product large enough to be
`sucient for both initial preclinical and clinical studies.
`The usual battery of toxicology studies for anticancer
`agents should be conducted unless there is adequate
`human safety experience. Since it is dicult to correlate
`
`179
`
`speci®c drug product components with pharmacologic
`action, attempts should be made early in the develop-
`ment scheme to control the manufacturing processes to
`produce consistent batches for subsequent preclinical
`and clinical study. Further e€orts should be made in the
`later stages of development to identify biologic assays
`which can be used to assure activity and as release
`speci®cations for the marketed product.
`Herbal products represent a specialized subset of al-
`ternative therapies, as there is often signi®cant human
`experience with their use. If there is a documented his-
`tory of use of the herbals or if these preparations are
`freely marketed in the United States, then no preclinical
`pharmacology or toxicology is required for initial trials
`using the marketed product. Submission of data on the
`traditional use, preparation of the product, and safety
`pro®le of any known components of the herbal prepa-
`ration for the IND is encouraged. When a product dif-
`ferent from the marketed version is intended for the
`clinical trial,
`information on the preparation of the
`product to be tested is import

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket