C. Erlichman M.D. OUTLINE Preclinical Development

1 / 90
About This Presentation
Title:

C. Erlichman M.D. OUTLINE Preclinical Development

Description:

C. Erlichman M.D. OUTLINE Preclinical Development Trials: Phase 1 Phase 2 Phase 3 Biomarkers in Cancer Treatment Trials Informed Consent and Regulations PRECLINICAL ... – PowerPoint PPT presentation

Number of Views:80
Avg rating:3.0/5.0

less

Transcript and Presenter's Notes

Title: C. Erlichman M.D. OUTLINE Preclinical Development


1
DEVELOPMENT OF NEW TREATMENTS IN CANCER
  • C. Erlichman M.D.

2
OUTLINE
  • Preclinical Development
  • Trials Phase 1 Phase 2 Phase 3
  • Biomarkers in Cancer Treatment Trials
  • Informed Consent and Regulations

3
PRECLINICAL STUDIES
4
LET ME SEE, WHICH DRUG SHOULD I TEST?
5
NCI Drug Development Stages
6
NCI Drug Development Stages
7
SCREENING FOR NEW DRUGS
  • Testing in tissue culture (in vitro)
  • 60 human cancer cell lines
  • patterns identified that suggest a new way of
    killing cancer cells
  • interesting agents tested in animal models

8
(No Transcript)
9
Advantages and Disadvantages of Cancer Cell Line
Drug Screens
  • Advantages
  • Tumor cell lines have identified active agents in
    clinical use
  • Identifies agents with antitumor activity in a
    cell type
  • May provide useful clue for clinical testing
  • Disadvantages
  • Mechanism of action not elucidated
  • Does not distinguish between nonspecific cell
    toxins and potentially useful agents

10
Preclinical Pharmacology
  • Development of Drug Plasma Method
  • Determine the extent of protein binding
  • Plasma elimination kinetics of i.v.
    administration
  • Other routes of administration such as oral, may
    be evaluated as well
  • The in vitro and in vivo metabolism is
    characterized using liver extracts

11
Preclinical Pharmacology
  • Preliminary (Range-Finding) Toxicity
  • For each drug, it is necessary to establish a
    maximum tolerated dose (MTD) and dose limiting
    toxicities (DLT) in both rodent and non-rodent
    species.
  • Testing in human tumor models growing in mice to
    determine whether the drug has activity
  • For each drug, the effect of treating animals
    with different tumors is evaluated looking at
    stopping tumors from growing and prolonging of
    the animals life
  • tumor types tested depends in part of the
    mechanism of action and where there is activity
    in tissue culture

12
IND Toxicology
  • Single or multiple daily dose schedules such as
    Dx1, q3hr x 3, q8hr x 15, q4D x 3, etc.
  • Up to twenty-eight days or more of repeated
    administration of drug to rodents and
    non-rodents.

13
IND Toxicology
  • Multiple cycle studies may be required if delayed
    or cumulative toxicity is anticipated.
  • Special studies such as cardiotoxicity,
    neurotoxicity evaluations, and immunotoxicity
    studies may be required as part of an existing
    study or in a separate study.

14
CANCER CLINICAL TRIALS
15
ELIGIBILITY CRITERIA
  • Primarily to ensure safety
  • To ensure some level of uniformity of the study
    population
  • To select a population of interest

16
COMMON ELIGIBILITY CRITERIA
  • Evidence of cancer
  • Age
  • Performance status
  • Adequate blood counts
  • Adequate liver function blood tests
  • Adequate kidney function
  • Reasonable life expectancy ? 3 months
  • Capable of giving informed consent

17
COMMON INELIGIBILITY CRITERIA
  • Has had therapy (XRT, Chemo) within 4 weeks
  • Pregnancy
  • Breast feeding
  • Prior treatments that may be similar
  • Recovery from surgery
  • Infections
  • Brain metastases
  • Cannot take oral medications

18
Phase I Trial Goals
  • 1. To determine MTD and phase II recommended dose
  • 2. To describe all toxicities
  • 3. To determine the clinical pharmacokinetics
    (PK)
  • 4. To report any responses
  • 5. To relate PK to toxicities and/or biologic
    effect
  • 6. To evaluate potential markers of biologic
    endpoints (pharmacodynamics)
  • 7. To relate PK to genetic polymorphisms

19
Pharmacokinetics Pharmacodynamics Pharmacogenetics
  • Pharmacokinetics
  • What the body does to the drug
  • AUC, clearance, t1/2, volume of distribution,
    Cmax, CSS, Tmax, renal clearance

20
Pharmacokinetics Pharmacodynamics Pharmacogenetics
  • Pharmacodynamics
  • What the drug does to the body
  • Toxicity Grade
  • Effect on ANC, platelets
  • Tumor response
  • Normal Tissue Surrogates
  • Effect on Imaging characteristics

21
Pharmacokinetics Pharmacodynamics Pharmacogenetics
  • Pharmacogenetics
  • Genetic variation in enzymes that metabolize
    drugs
  • Single nucleotide polymorphisms (SNPs)
  • Functional SNPs
  • Differences between ethnic groups
  • Frequency could be common or relatively rare
  • Not analyzed for cancer risk, screening, or
    prevention

22
Phase I Trial Design Considerations
  • Starting dose
  • 1/10 LD10 in mice provided the dose lt TLD (toxic
    low dose) in dog
  • If toxic in dog, then start with1/3 TLD in dog

23
Phase I Trial Design Considerations
  • Standard cohort-escalation design
  • 3 patients per dose level
  • If 2 or 3 DLTs stop escalation
  • If 1 DLT add 3 patients and if no further DLT
    continue dose escalation, i.e. 1/6 DLT
  • MTD is 1 dose level lower

24
ASSESSMENT OF DRUG TOXICITY (SIDE EFFECTS)
25
TOXICITY GRADING ATTRIBUTION
  • NCI common toxicity criteria (CTC v3.0)
  • Not related
  • Possible related
  • Probably related
  • Definitely related

26
Example of CTC Toxicity Tables
27
PHASE 2 AND 3 STUDY GOALS
  • Response Rate What percent of patients have
    significant tumor shrinkage
  • Time to Progression The time it takes for the
    tumor to grow by a certain amount after starting
    the study
  • Disease Free Survival The time it takes for the
    tumor to come back after starting the study
  • Overall Survival The time patients live after
    starting the study

28
PHASE 2 CLINICAL TRIALS
  • Initiated when a safe dose and schedule define in
    phase I
  • Performed in specific tumor types
  • Tumor types selected for testing depending on a
    variety of criteria
  • Overall purpose is to determine whether there is
    any clinically meaningful activity

29
PHASE 2 CLINICAL TRIALS
  • Criteria for tumor types to study
  • hint of effect in phase I
  • specific target is present in that cancer
  • a clinical need is identified
  • competitors in the market place
  • potential market size

30
PHASE 2 CLINICAL TRIALS GOALS
  • To determine the response rate, time to
    progression and survival
  • To determine the frequency and spectrum of all
    toxicities
  • To relate potential markers of biologic endpoints
    to clinical effect
  • To relate pharmacogenetics to toxicity and
    efficacy

31
Phase I vs PHASE 2 CLINICAL TRIALS
  • Phase I trials include 20-25 patients with many
    tumor types and patients are treated at different
    doses
  • Phase 2 trials can include from 14 to 100
    patients with one tumor type treated with one dose

32
PHASE 2 CLINICAL TRIALS
  • If a phase 2 trial meets its objective then phase
    3 trials are needed
  • However, phase 2 trial results can be used for
    accelerated approval by FDA
  • This needs to be established in consultation with
    FDA before the plan for the trial is finalized
    and will still require a phase III trial

33
PHASE 3 CLINICAL TRIALS
  • Definitive comparative trials
  • Requires a randomization
  • Rarely blinded in Cancer trials
  • Primary endpoints can include response rate, time
    to progression, overall survival and disease-free
    survival

34
PHASE 3 CLINICAL TRIALS
  • As follow-up on patients may be long results can
    take 5 and 10 years to be realized
  • Size of the study can vary from a few hundred
    patients to thousands

35
Molecularly Targeted Therapies
  • A new paradigm for cancer treatment
  • Cancer as a chronic disease like diabetes,
    hypertension
  • Although cure is still the ultimate goal,
    control can achieve valuable benefit for the
    patient
  • Treatment are less toxic and chronic

36
Natural History of Cancer
Cellular Dedifferentiation
Growth Dysregulation
Loss of Apoptosis
Invasion and Metastasis
Unlimited Cell Division
Angiogenesis
37
Molecular Events in Cancer
Dysregulation of Growth Factors or Receptors
Aberrant Signal Transduction
Secretion of Autocrine Growth Factors
Secretion of Angiogenic Growth Factors
Secretion of Matrix Metalloproteinases
Expression of Oncogenes Loss of Tumor Suppressor
Genes
38
Receptor Tyrosine Kinases Overexpressed in Breast
Cancer
Extracellular
region
Flt-1
Flk-1
a
ß
a
a
N-Flg
ß
ß
TRK-B
(TK-)
TRK family
FGF receptor
Tie
Insulin
Ltk
EPH/ECK
Ret
Ros/Sev
Met/Sea
Axl/Ark
family
PDGF receptor
receptor
Family
Trk
EGF
family
Family
family
family
family
Trk-B
receptor
Flg
Trk-C
family
c-Ros
Met (HGFR)
Eph
Axl
a
N-Flg
PDGFR-
INS.R
Sevenless
c-Sea?
Eck
Ark
Bek
IGF1R
PDGFR-ß
EGFR
Elk
FGFR-3
c-Fms(CSF1-R)
Neu
Eek
FGFR-4
IgG-like domain
c-Kit(SGFR)
ErbB3
Hek
Cysteine-rich box
Flt-1
EGF-like repeats
Flk-1
Fn type III repeats
Flk-2 (Flt-3)
Transmembrane helix
Intracellular
Flt-4
Plasma membrane
region
Catalytic domain
39
Molecularly Targeted Therapies
  • Drugs that target receptor tyrosine kinases
    Gleevec, Tarceva
  • Antibodies that target receptors Herceptin,
    Erbitux
  • Antibodies that target receptor ligands - Avastin

40
Anti-EGF Agents (Antibodies)
  • MoAb
  • Erbitux
  • ICR62
  • mAb425
  • Ior egf/r3
  • Pantuzamab
  • h-R3
  • EMD72000
  • Bispecific Antibodies
  • MDX-447
  • Toxin-Linked Conjugates
  • ScFV (14E1)-ETA
  • ScFV (225)-ETA
  • MRI (Fv)-PE38
  • TP40
  • DAB389-EGF

41
Anti-EGF Agents (Drugs)
  • AG1478
  • CGP59326
  • CI1033
  • EKB569
  • IRESSA
  • TARVECA
  • LAPATINIB
  • PKI-166
  • PD158780

42
Tumor AngiogenesisAngiogenic Balance or Switch
VEGF, bFGF, TGF-?, integrins, MMP, IL8, hypoxia,
NO (etc)
Angiostatin, endostatin, interferons, TIMP (etc)
Growing capillaries
Metastasis
43
VEGF Ligands and Receptors
VEGFR-2 (Flk-1/KDR)
VEGFR-3 (Flt-4)
VEGFR-1 (Flt-1)
Angiogenesis
Angiogenesis
Lymphangiogenesis
Lymphangiogenesis
PlGF Placental growth factor
44
Anti-Angiogenesis Agents
  • Tyrosine Kinase Inhibitors
  • Sorefinib
  • Sunitinib
  • CHIR-258
  • AEE788
  • BIBF1120
  • AZD 2171
  • Biologic Agents
  • Bevacizumab
  • VEGFR-TRAP

45
Biomarkers in Cancer Clinical Trials
  • Should we treat only those patients that express
    the target?
  • Do we need to treat at doses that cause some side
    effects like with chemotherapy?
  • Should we measure the benefit in the same way
    e.g. response rate?

46
Biomarkers in Cancer Clinical Trials
  • Should the agent be highly specific or affect
    several target?
  • If the latter then how many?
  • Should these agents be used in combination with
    chemotherapy?
  • Should these agents be combined with each other?

47
By Liz Szabo, USA TODAY
48
Can we improve the effectiveness and/or decrease
the risk associated with cancer treatment while
keeping costs under control?
49
Biomarkers for Target Effect
  • Presence of the target eg ER, PR, HER2
  • Effect of treatment on plasma tumor marker eg
    CA125, PSA, aFP.
  • Effect of treatment on a downstream molecule eg
    phospho-p70S6K with CCI-779 or RAD001
  • Depletion of circulating tumor cells after
    treatment
  • Decrease in metabolic imaging after treatment

50
Types of Biomarkers
  • Biologic Fluids plasma, serum, urine, CSF,
    effusions
  • Tissue Normal or tumor
  • Proteomic, Genomic, DNA
  • Imaging
  • PET scanning 18FDG, 18FLT, H2O15
  • MRI
  • CT perfusion

51
IRESSA
Albanell. J Clin Oncol 20(1)7110-124, 2001.
52
IRESSA
Albanell. J Clin Oncol 20(1)7110-124, 2001.
53
IRESSA
Albanell. J Clin Oncol 20(1)7110-124, 2001.
54
Therapy of Patients with Metastatic Colon Cancer
  • How do we know if the treatment is working?
  • Wait 2 months and expose the patient to the side
    effects
  • Does a test that predicts the effectiveness
  • Blood test
  • Imaging

55
GIST PET Shows a complete response before and
after 5 days of Gleevec
56
FDG-PET predicts response to chemotherapy in
NSCLC. Median PFS and OS are significantly longer
for responders than nonresponders (163 versus 54
days and 252 versus 151 days, respectively).
Kelloff, G. J. et al. Clin Cancer Res
2005112785-2808
57
Circulating tumor cells
  • Peripheral blood epithelial cells shed from tumor
    surface of cancer patients
  • Defined by phenotypic characteristics
  • Expression of cytokeratin or mucin markers such
    as CK18, CK19, and MUC-1
  • Phenotypically similar to primary and/or
    metastatic tumor cells

Fehm T, et al. Clin Cancer Res. 200282073-2084.
58
Detection Methods
  • Direct methods (enrichment/detection)
  • Immunomagnetic
  • Immunohistochemistry (IHC)
  • PCR analysis
  • Automated fluorescent methods

59
Prognostic and Predictive Value of CTCs in MBC
  • Hypothesis
  • Measurement of CTCs in metastatic breast cancer
    may
  • Identify aggressive disease (prognostic value)
  • Provide early determination of treatment
    efficacy/benefit (predictive value)

60
Prognostic Value of Baseline CTC Counts
Overall Survival
100
90
80
70
60
Log rank P lt .0001
Probability of Progression-Free Survival
21.9 months
50
10.9months
40
30
20
n 87 (49)
10
0
0
2
4
6
8
10
12
14
16
18
22
24
26
28
30
20
Time From Baseline (Months)

Cristofanilli M, et al. N Engl J Med.
2004351781-791.
61
Change in CTC Count During Therapy Predicts
Overall Survival
1
lt5 CTCs at all time points
83 (47)
2
gt 5 at baseline and lt 5 CTC at last draw
38 (21)
3 lt 5 at Early Draw and gt 5 CTC at last
draw 17 (10)
100
4
gt 5 CTCs at all time points
39 (22)
90
1
vs
2
P .3188
80
1
vs
3
P .0014
70
1
vs
4
P lt .0001
60
Probability of Survival
2
50
1
40
2
vs
3
P .0397
30
20
2
vs
4
P lt .0001
10
3
vs
4
P .0051
4
3
0
0
2
6
8
10
12
14
16
18
22
24
26
28
20
4
30
Time From Baseline (Months)

Cristofanilli M, et al. ASCO
2005. Abstract 524.
62
(No Transcript)
63
(No Transcript)
64
(No Transcript)
65
(No Transcript)
66
(No Transcript)
67
Tamoxifen metabolic pathway
68
Tamoxifen Pharmacogenetics(Goetz, Ingle,
Weishilboum, Flockhart)
  • Does CYP2D6 genetic variation affect the clinical
    outcomes of women receiving tamoxifen?
  • Do women who are co-prescribed medications which
    inhibit CYP2D6 have a higher risk of breast
    cancer relapse?

69
CYP2D6 Gene
  • Encompasses nine exons with an open reading frame
    of 1383 bp coding for 461 amino acids. Highly
    polymorphic
  • At least 46 major polymorphic alleles with 4
    well-defined phenotypes
  • Poor metabolizers (PM)
  • Intermediate metabolizers (IM)
  • Extensive metabolizers (EM)
  • Ultra-rapid metabolizers (UM)

70
CYP2D6 Gene
  • CYP2D6 PM 7-10 of Caucasians lack functional
    CYP2D6
  • CYP2D6 4 Most common variant leading to null
    CYP2D6 enzyme in Caucasians (accounts for 75 of
    the Caucasian PM)

71
NCCTG 89-30-52
RANDOMIZATION
Postmenopausal women Early ER breast cancer
5 years of Tamoxifen
5 yrs of Tamoxifen 1 yr of Fluoxymesterone
(10 mg po bid)
541 women accrued
5 years total therapy
72
NCCTG 89-30-52
  • Median follow-up of 12 years
  • Accrual completed in April 1995
  • No difference between the arms in terms of
    primary endpoint (breast cancer relapse)
  • Ingle J, Suman V, Mailliard J, et al Breast
    Cancer Res Treat (In Press)

73
Tamoxifen only arm
  • 230/257 eligible patients--Formalin fixed
    paraffin-embedded tumor blocks
  • Three 10 micron sections
  • DNA extracted
  • Genotyping
  • CYP2D6 (4) (n190)
  • CYP3A5 (3)
  • SULT1A1 (2)

74
Relapse-free Time
CYP2D6 Wt/Wt
CYP2D6 4/Wt
CYP2D6 4/4

P0.030
Years after randomization
CP1193836-1
Goetz et al J Clin Oncol. 200523(36)9312-8.
75
Relapse-free Survival
CYP2D6 WT/WT
CYP2D6 4/WT

CYP2D6 4/4
P0.020
Years after randomization
Goetz et al J Clin Oncol. 200523(36)9312-8.
CP1193836-2
76
Informed Consent and Regulations
77
Ethics of Phase I Trials
  • No known benefit
  • Risk of toxicity is unknown
  • Patients expectations are high
  • Patients discount risk of toxicity

78
Ethics of Phase I Trials
  • Patient assessment of benefit
  • Phase I 59.8
  • Standard 36.8
  • Patient assessment of toxicity
  • Phase I 29.8
  • Standard 45.6
  • Based on analyses of multiple phase I trials
  • Benefit (Response) 5
  • Toxicity (Death) 0.5

79
INFORMED CONSENT
80
Consent Form Excerpts
  • Why is this study being done?
  • This study is being done to
  • Learn the highest safe dose of the
    investigational drug 17-AAG when it is given over
    1 hour once each week for 3 out of every 4 weeks
    to patients with advanced cancer.
  • Learn the side effects of 17-AAG and how the body
    responds to and removes 17-AAG.

81
Consent Form Excerpts
82
Consent Form Excerpts
83
Consent Form Excerpts
  • Are there benefits to taking part in this study?
  • No help can be promised by taking part in this
    study, and the chance of help from this study
    cannot be accurately predicted.

84
Consent Form Excerpts
85
Consent Form Excerpts
  • During the first month of this study, the
    following additional tests and procedures will be
    done to test the amount of 17-AAG in your body
    and to learn what effect it has on your body.
    These will be done at no additional cost to you
    and/or your health plan and include
  • Nineteen (19) blood samples (totaling about 1¼
    cup) will be collected.
  • Before and at one day after your first and third
    weekly 17-AAG treatments, samples of tissue will
    be taken by gently scraping the inside of your
    cheeks or mouth with a tongue depressor.
  • You will need to save your urine for 24 hours
    before and after your first 17-AAG treatment.
  • If you have a tumor that can be safely and easily
    biopsied, you may be asked to have two additional
    biopsies. One before you start and the other one
    day after the start of your treatment.

86
Consent Form Excerpts (HIPAA)
  • Authorization To Use And Disclose Protected
    Health Information
  • By signing this form, you authorize Mayo Clinic
    Rochester and the investigators to use and
    disclose any information created or collected in
    the course of your participation in this research
    protocol.
  • This information may include information relating
    to sexually transmitted disease, acquired
    immunodeficiency syndrome (AIDS), or human
    immunodeficiency virus (HIV). It may also include
    information relating to behavioral or mental
    health services or treatment and treatment for
    substance abuse.

87
Consent Forms in Phase I Oncology Trials
  • S. Horng et al NEJM 2002 347(26)2134-2140
  • never promise benefit
  • rarely mention cure
  • communicate risk and its unpredictability
  • do not contribute to patients expectation of
    benefit

88
ADHERENCE TO STUDY PROTOCOL IS CRITICAL
89
COMPLIANCE WITH REGULATIONS
  • NCI regulations
  • Cancer Center
  • Cooperative groups
  • FDA regulation
  • OHRP regulations as interpreted by IRB

90
INVESTIGATOR OBLIGATIONS
  • Ensure informed consent is obtained
  • Report serious adverse events
  • Report adverse events
  • Data monitoring safety plan
  • Data safety monitoring committee
  • Protocol compliance
  • Regular auditing of cases on study

91
QUESTIONS ARE WELCOME
Write a Comment
User Comments (0)