SlideShare a Scribd company logo
1 of 72
Evaluation of anticancer agents
- Dr Anup Thorat
- Guide : Dr Sandhya Kamat
History
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 2
History
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 3
Louise Goodman
Sidney Farber
Father of modern
chemotherapy
Alfred Gilman
Need for Novel Anticancer Agents
• Multidrug resistance.
• Long-term treatment with cancer drugs a/w severe side
effects.
• Cytotoxic drugs have the potential to be very harmful to the
body unless they are very specific to cancer cells.
• New drugs - more selective for tumour cells.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 4
Pre clinical evaluation
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 5
IN VITRO IN VIVO
In vitro cytotoxicity studies:
• 1990 : NCI-60 screen
• Cytotoxicity assays on panel of human cancer cell lines
– MTT-assay
– SRB- assay
– 3H-thymidine uptake assay
– Fluorescence Dye exclusion tests
– Clonogenic assays
– Cell counting assay
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 6
In vitro cytotoxicity studies:
Advantages
• Reduce the usage of animals.
• Less time consuming,
• Cost effective &
• Easy to manage
• Able to process a larger number of
compounds quickly with minimum
quantity.
• Range of concentrations used are
comparable to that expected for in vivo
studies
Disadvantages:
• Difficulty in maintaining of cultures.
• Show negative results for the
compounds which gets activated after
body metabolism and vice versa.
• Impossible to ascertain the
Pharmacokinetics .
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 7
Cell lines
• Tumor cell lines derived from several cancer types
– lung, colon, melanoma, renal, ovarian, brain, and leukemia.
• The lines are prepared and cryopreserved using DMSO(di
methyl sulfoxide )
• THAWING OF THE CELLS : Rapid thawing of frozen ampoule in
water bath until it gets liquefied.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 8
Cell lines
• The solution is centrifuged with saline for 10 mins to remove
the DMSO
• The saline is discarded and aliquot is taken for cell counting,
cell viability and for sub culturing.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 9
CELL PROPERTIES & ASSAY
PROPERTIES ASSAY
ENZYMATIC PROPERTY MTT ASSAY
MEMBRANE INTEGRITY DYE EXCLUSION TEST
DNA CONTENT/REPLICATION STATUS 3H-THYMIDINE UPTAKE & FLUORESCENT ASSAY
PROTEIN CONTENT SRB ASSAY
COLONY FORMING POTENTIAL CLONOGENIC ASSAY
CELL DIVISION CELL COUNTING ASSAY
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 10
Microculture Tetrazolium Test(MTT Assay)
• A quantitative colorimetric assay
• Measures cellular growth, cell survival and cell proliferation
based on the ability of living cells.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 11
MTT
(3-(4,5-
Dimethylthiazol-2-yl)-
2,5-
diphenyltetrazolium
bromide)
Formazan
mitochondrial dehydrogenase
in living cells only
MTT Assay - Procedure
• Cells from particular cell lines in log phase of growth are
trypsinised. Check the cell viability through haemocytometer .
• Adjust to appropriate density in suitable medium and inoculate in
multiwell plates.
• Cells are treated with various conc. of test compounds & the plate
incubated at 37 °C in 5% CO2 /95% humidified air (1-4 d)
• Cultures are taken out and 10 μl of MTT dye is added (5 mg/ml)
into each well and incubated for 4hrs
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 12
MTT Assay
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 13
MTT Assay - Procedure
• The plate is centrifuged, supernatant discarded & precipitated
formazan salt is dissolved in 100 μl of isopropanol/DMSO
• The plate samples are read at 570 nm microtiter plate reader
IC50 of drugs can be determined by counting the viable cells
% Cell viability = Absorbance of treated cells x 100
(%MTT reduction) Absorbance of untreated cells
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 14
Microtiter plate reader
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 15
SRB (SULPHORHADAMINE B) ASSAY
• Measures whole protein content which is proportional to the
cell number.
• SRB – a bright pink anionic protein staining dye that binds to
the basic amino acids of the cellular proteins.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 16
SRB ASSAY - Procedure
• Cell lines are counted, cultured & inoculated in 96 well plates.
• After incubation with different concentrations of test
compounds, the cell cultures are stained with SRB dye.
• Washing with acetic acid removes the unbound dye and the
protein bounded dye is extracted using triss base
• Optical density is determined by 96-well plate reader
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 17
3H-Thymidine uptake assay
• Provides information about tumour kinetics, ploidy status of
the cells.
Procedure:
1. Tumor cell suspensions are exposed to the
drug for 5 days.
2. Radio-labeled 3H-thymidine is added
3. Replicating cells will incorporate 3H-thymidine into their DNA
4. Determined by autoradiography or liquid scintillation.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 18
FLUORESCENCE ASSAY
• Cells are exposed to fluorescent labeled precursors after drug
exposure.
• Replicating cells will incorporate the dye into their DNA.
• Resulting fluorescence is measured by flow cytometry /
fluorescent microscopy .
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 19
Dye Exclusion Test
• This assay is relayed on the structural integrity of the cells.
• Dyes used : Trypan blue, Eosin, or Nigrosin
• Live cells possess intact cell membranes that exclude the dye,
whereas dead cells having lost membrane integrity take up the
dyes.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 20
Dye Exclusion Test - Procedure
• Cells are incubated with different concentrations of test compounds for
4 days.
• Dead cells are stained with dye colour.
• Specimen is centrifuged & collected in microscopic slides.
• Live cells – stained with hematoxylin eosin.
• Tumor cell cytotoxicity is compared with control – duck erythrocytes.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 21
Trypan blue dye exclusion test
Live
cell
Dead
cell
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 22
CLONOGENIC ASSAY
• Measures the growth inhibition.
• Important for the drugs that act by arresting the cells at checkpoints in the
cell cycle
Procedure:
1. Single cell suspensions are exposed to anticancer agents to be tested.
2. Suspensions are rinsed and plated in a semisolid medium.
3. After 14 to 28 days, some cells will having undergone several division
form tumour colonies which can be quantified in a visual or semi-
automated fashion.
4. No of colonies from treated cells is compared with untreated colonies.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 23
CLONOGENIC ASSAY
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 24
Preclinical Toxicity Studies
• Aimed at predicting
(a) Safe starting dose & dosage regimen for human clinical trials(P1)
(b) The toxicities of the compound, &
(c) The likely severity and reversibility of drug toxicities.
• Regulatory requirement : Two acute preclinical toxicity studies
1. Rodent (mice) - single- and multiple-dose lethality studies.
2. Non rodent (dogs) - single- and multiple-dose confirmatory toxicity.
• Cytotoxic & non cyotoxic drugs
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 26
Acute Toxicity Studies
• First mouse given a single injection (IP, IV, SC, IM or PO) of 400 mg/kg (or
lower if the compound is extremely potent)
• Second mouse - 200 mg/kg &
• Third mouse - 100 mg/kg
• The mice are observed for a period of 2 weeks & sacrificed if there are
signs of significant toxicity
• If all 3 mice must be sacrificed, the next 3 dose levels (50, 35 and 12.5
mg/kg) are tested in a similar manner
• This process is repeated until a tolerated dose is found designated as MTD.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 27
Preclinical Toxicity Studies
• To determine the phase I entry dose of a cytotoxic anticancer
agent, the dose levels that are lethal to 10, 50, and 90% of mice
(LD10, LD50, & LD90) is determined by the same route of
administration.
The projected phase I entry dose is usually 1/10th of the LD10
• A robust toxicity study needed to avoid lengthy phase I trials as
well as severe drug toxicities.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 28
Why Mice?
• Mice are small, easy to handle.
• Short generation time & accelerated lifespan,manageable
costs, space, and time.
• Striking similarity to humans in anatomy, physiology, and
genetics.
• Over 95% of the mouse genome is similar to humans.
• Many of the genes responsible for complex diseases are shared
between mice and humans.
• Mouse genome can be directly manipulated.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 29
Tumour Models
1. Carcinogen induced models
2. Viral infection models
3. Transplantation Models
– Murine
– Human
4. Genetically Engineered Mouse Models
5. In vivo hollow fibre assay
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 30
CHEMICAL CARCINOGEN MODEL
DMBA induced mouse skin papillomas
• Two stage experimental carcinogenesis
– Initiator – DMBA (dimethylbenz[a]anthracene),
– Promotor – TPA. (12-O-tetradecanoyl-phorbol-13-acetate)
• Mice : Single dose – 2.5 µg of DMBA f/b 5 to 10 μg of TPA in
0.2 ml of acetone twice weekly.
• Papilloma begins to appear after 8 to 10 wks - Tumor incidence
& multiplicity of treatment group is compared with DMBA
control group
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 31
Mouse skin papillomas
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 32
MNU INDUCED RAT MAMMARY GLAND CA
• Induces hormone dependent tumors.
• Single i.v of 50 mg/kg of methylnitrosourea - 50 days old SD
rats.
• Adenocarcinoma will be produced within 180 days of post –
carcinogen – 75 to 95%
• Reduction in tumor size is compared
• Drawback – cannot detect inhibition of carcinogen activation.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 33
RAT MAMMARY GLAND CA
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 34
DMBA induced rat mammary gland ca
• Sprague dawley rats
• Intragastric injection of 12 mg / kg of DMBA at 50 days of age
• Cancer appears within 120 days – 80 to 100%
• Detect the drugs inhibiting carcinogen activation.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 35
MNU induced tracheal squamous cell ca
• Hamster
• 5% of MNU in normal saline is given once a week for 15 wks by
catheter.
• Within 6 months – 40 to 50%
• Test drug efficacy is measured by comparing with carcinogen
control group
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 36
DEN induced lung adenocarcinoma in Hamster
• 17.8mg DEN/kg body wt twice weekly by S.C inj for 20 weeks
starting at age 7 to 8 weeks.
• Usually produces tracheal tumors in 90-100% and lung tumors
in 40-50% of male syrian hamster.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 37
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 38
Cancer site Cancer Type Species Carcinogen
Colon Adenocarcinomas Rat AOM (azoxymethane)
Prostate Adenocarcinomas Rat MNU (methylnitrosourea)
Esophagus Squamous cell
carcinoma
Rat NMBA(N-nitroso-
methylbenzylamine)
Breast Adenocarcinoma Mice NMU (N-Nitroso-N-
methylurea)
Viral infection models
• Mouse Mammary Tumor Virus (MMTV) was the first mouse
virus, isolated at Jackson labs as the “non-chromosomal
factor” that caused mammary tumors in the C3H strain of
mice.
• Some viruses cause cancer via random integration in certain
cells
• Some viruses carry cellular oncogenes
– Abelson murine leukemia virus – Abl
– Moloneymurine sarcoma virus – Raf
• Engineered viruses now used routinely in the laboratory to
induce cancer.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 39
Transplantation Models
• Tumor cells or tissues (mouse or human) transplanted into a
host mouse.
• Ectopic – Implanted into a different organ than the original
(typically subcutaneous or kidney capsule)
• Orthotopic – Implanted into the analogous organ of the
original tumor.
• Advantages :
– Typically cheap, fast & easy to use.
– Not covered by patents
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 40
Transplantation Models
• Disadvantages:
– Histopathology cannot be exactly correlated to human tumors
– Lack of step-wise progression through pre-neoplastic stages
– Evolution of tumor cells during passaging
– Requirement for angiogenesis to support the newly transplanted
tumor
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 41
Transplantation Models : Human Tumor Xenografts
• Athymic “nude”mice developed in 1960’s
• Mutation in nu gene on chromosome 11
• Phenotype: retarded growth, low fertility,
no fur, immunocompromised
– Lack thymus gland, T-cell immunity
• First human tumor xenograft of colon adenocarcinoma by Rygaard & Poulson,
1969
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 42
Xenograft Sites
• Subcutaneous tumor (NCI method of choice) with IP drug
administration
• Intraperitoneal
• Intracranial
• Intrasplenic
• Renal subcapsule
• Site-specific (orthotopic) organ inoculation
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 43
Xenograft Study Endpoints
• Toxicity Endpoints:
– Drug related death
– Net animal weight loss
• Efficacy Endpoints:
– Tumor weight change
– Treated/control survival ratio
– Tumor growth assay (corrected for tumor doubling time)
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 44
Xenograft Tumor Weight Change
• Tumor weight change ratio (used by the NCI in xenograft
evaluation)
• Defined as: treated/control x 100%
• Tumor weight in mg = (a x b2)/2
– a = tumor length
– b = tumor width
• T/C < 40-50% is considered significant
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 45
Human Tumor Xenografts
Advantages
• Many different human tumor cell
lines transplantable
• Wide representation of most
human solid tumors
• Good correlation with drug
regimens active in human lung,
colon, breast, and melanoma
cancers
Disadvantages
• Brain tumors difficult to model
• Different biological behavior,
metastases rare
– Survival not an ideal endpoint:
death from bulk of tumor, not
invasion
• Shorter doubling times than
original growth in human
• Difficult to maintain animals due
to infection risks
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 46
Transplantation Models : Cell line based allografts
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 47
Transplantation Models : Cell line based allografts
Advantages
• Very easy to maintain cell lines
indefinitely;
• Very fast to generate large
numbers of tumors
• Relative homogeneity of tumors
makes it easy to detect
differences
• Comparatively cheap
• Intact immune system
Disadvantages
• Mouse cells rather than human
• Inaccurate histopathology
compared to human tumors
• Evolution of tumor cells during
culture
• As it involves murine CA, ability
to predict response to therapy in
humans is controversial.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 48
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 49
In Vivo Hollow Fibre Assay
• In vivo screening tool implemented in 1995 by NCI
• 12 human tumor cell lines (lung, breast, colon, melanoma,
ovary, and glioma
• Cells suspended into hollow polyvinylidene fluoride fibers
implanted IP or SC in lab mice
• After in vivo drug treatment, fibers are removed and analyzed
in vitro
• Antitumor (growth inhibitory) activity assessed
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 50
In Vivo Hollow Fibre Assay
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 51
In Vivo Hollow Fibre Assay
Subcutaneous Hollow
Fibre implants
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 52
Clinical Evaluation
Clinical Evaluation
• The most reliable method for demonstrating efficacy is to
show a statistically significant improvement in a clinically
meaningful endpoint in blinded randomized controlled trials
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 54
Regulatory considerations
1. Regular Approval
– Longstanding route of drug approval based on the demonstration of
clinical benefit
2. Accelerated Approval
– Use of a surrogate endpoint that is reasonably likely to predict
benefit.
– Serious or life-threatening diseases
– Improvement over available therapy
– No existing therapy
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 55
Endpoints
• 1970s - Objective Response Rate (ORR)
• 1980s - improvement in survival, quality of life (QOL), physical
functioning, tumour related symptoms.
• Three kinds of end points have been used:
1. Overall Survival
2. Tumor assessment endpoints e.g. ORR,DFS, PFS,TTP, TTF; and
3. Symptom assessment endpoints e.g. palliation of side effects, QOL
scores.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 56
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 57
Endpoint Regulatory
Evidence
Study Design Advantages Disadvantages
Overall
Survival
(OS)
Clinical benefit
for regular
approval
Randomized
studies essential
Blinding not
essential
1)Universally accepted
direct measure of
benefit
2)Easily & Precisely
measured
1)May involve larger studies
2)May be affected by crossover
therapy and sequential therapy
3)Includes non cancer deaths
Symptom
Endpoints
(patient
reported
Outcomes)
Clinical benefit
for regular
approval
Randomized
blinded studies
1)Patient perspective of
direct clinical benefit
1)Blinding is often difficult
2)Data are frequently missing or
incomplete
3)Clinical significance of small changes
is unknown
4)Multiple analyses
5)Lack of validated instruments
Objective
Response
Rate
(ORR)
Surrogate for
accelerated
Approval
or
regular
Single-arm or
randomized
studies can be
used
Blinding
1)Can be assessed in
single-arm studies
2)Assessed earlier and in
smaller studies
compared with survival
1)Not a direct measure of
benefit
2)Not a comprehensive
measure of drug activity
3)Only a subset of patients
Study Designs
Single-Arm Studies
• No available therapy
• ORR (objective response rate) and response duration in single-arm
studies can be a substantial evidence supporting accelerated
approval.
• Primary efficacy measure : Proportion of patients who achieve a
complete or partial response to the treatment.
• Design eliminates truly ineffective therapy
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 59
Single-Arm Studies
• Challenges :
1. Do not adequately characterize time-to-event endpoints (survival,
TTP, PFS)
2. Because of variability in the natural history of cancer, a randomized
study is necessary to evaluate time-to-event endpoints.
3. Inability to predict comparative performance vis-a`-vis the then-
available best possible, standard-of-care therapeutic option.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 60
Non-inferiority Studies
• Rely on historical data to establish the active control’s treatment
effect size & constancy assumption
• Estimated size of the active-control’s treatment effect should be
based on a comprehensive meta-analysis of historical studies
• Challenges :
1. the estimation of active-control effect and the determination of amount
of effect (NI margin) to be retained. Usually large sample sizes.
2. Subsequent therapies and crossover to the active-control arm can
confound any NI analysis
3. NI trials with endpoints other than survival are problematic
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 61
Clinical Trial Phases
• What is the dose and schedule? Phase I
• Is it active?
– spectrum of anticancer activity ? Phase II
– is it better than standard therapy? Phase III
• Is it safe? Phase I to IV
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 62
Phase 0 : Microdosing
• The FIH dose : significant safety risk for the patient volunteers.
• Less than1/100th of the dose calculated to yield a pharmacological
effect of the test substance to a maximum dose of less than 100 µg.
• Advantages:
– Mitigate FIH dose risk,
– Gather early pharmacokinetic data (bioavailability, clearance, elimination
rate), and
– Increased efficiency of drug development.
• Highly sensitive analytical methods : PET, LCMS, accelerated mass
spectroscopy(AMS)
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 63
Phase 1
• Usually conducted in patients, rather than healthy volunteers.
• This adds to the challenges :
1. Recruitment of tumor-specific patient volunteers
2. The recruited volunteers may be in the advanced stages of the
disease - refractory to the currently available standard-of-care
treatment options - cost escalation.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 64
Dose Escalation Studies
• Phase 1 entry dose is obtained from preclinical toxicity studies
• Endpoints :
– Along with MTD, info on clinical toxicity, pharmacokinetics, and
preliminary antitumor activity is obtained.
– Also for cytotoxic agents, dose-related toxicity is regarded as a
surrogate for efficacy.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 65
Progressively escalating
doses
MTD
Dose Escalation Studies
• Modified Fibonacci search :
1. Safe starting dose(d) = 1/10th of the LD10
2. Enroll patients in cohorts of three, and escalate the dose according
to a modified Fibonacci sequence
3. Higher escalation steps have decreasing relative increments -100,
65, 52, 40, 29% & thereafter 33% increases over the previous dose.
4. Ethical issues : substantial numbers of patients are treated at
nontherapeutic doses
5. Efficiency issues : Lengthy trials, time & cost incurred.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 66
Dose escalation : Other Methods
• Pharmacologically guided dose escalation (PGDE)
– Using the preclinical toxicology data to rapidly escalate doses to a
target area under the curve (AUC) value obtained from murine
pharmacokinetic data.
• Non-pharmacokinetic statistical modeling approaches :
– Statistical approaches model the dose–toxicity relationship as a
sigmoidal curve to predict the MTD. The toxicity is then evaluated &
actual MTD found by rapid dose titration.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 67
Ethical Challenges In Phase I
1. The paucity of benefits with substantial risks
2. Informed consent
• Majority of participants are treated at doses that cannot produce
responses in human tumors.
• The objections based on informed consent are deficiencies of
disclosure, understanding, and voluntariness
• Vulnerable group – Terminally ill patient
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 68
Phase II and III Clinical Trials
• Phase II studies are carried out in a small group of patients with a
specific tumor type to
– Determine anticancer efficacy &
– To define the therapeutic window of the compound
• Any arm may be terminated early because of discouraging results,
and the response rate of each arm is assessed separately against a
historical control rate with definable α and β error probabilities
• Phase III trials are conducted in a much greater number of patient
volunteers of the selected tumor type with prospective and
randomized evaluation against standard of care.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 69
Phase II and III Clinical Trials
• Embedding a randomized phase II study within a phase III
study
– Patients who progress on one of the arms of Phase II would then be
randomly assigned to arms E or C of phase III & and
– Their survival data would be combined in a stratified fashion with
data from patients directly assigned to arms C or E for the phase III
assessment.
– This design allows untreated patients to be enrolled onto the phase II
study, while assuring them a more established therapy if they
progress
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 70
Phase II and III Clinical Trials
• Phase II studies act as a screen of antitumor efficacy to select
the most promising agents to enter the pivotal phase III clinical
trials.
• A Phase II study should efficiently eliminate truly ineffective
therapy & reliably indicate whether subsequent phase III
testing is warranted.
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 71
5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 72

More Related Content

What's hot

Screening models on aphrodisiac agents
Screening models on aphrodisiac agentsScreening models on aphrodisiac agents
Screening models on aphrodisiac agentsJaineel Dharod
 
Hepatoprotective screening methods
Hepatoprotective screening methodsHepatoprotective screening methods
Hepatoprotective screening methodsShobhiniChandel
 
screening of anti-fertility agent
screening of anti-fertility agentscreening of anti-fertility agent
screening of anti-fertility agentJaineel Dharod
 
Screening of antipyretic drugs
Screening of antipyretic drugsScreening of antipyretic drugs
Screening of antipyretic drugsSindhoora Shetty
 
Screening models for aphrodisiac agents and anti fertility agents
Screening models for aphrodisiac agents and anti fertility agentsScreening models for aphrodisiac agents and anti fertility agents
Screening models for aphrodisiac agents and anti fertility agentsCh. Bhargava krishna
 
Preclinical screening of anti fertility agents
Preclinical screening of anti fertility agentsPreclinical screening of anti fertility agents
Preclinical screening of anti fertility agentsNaveen K L
 
Evaluation methods of anti-asthmatics
Evaluation methods of anti-asthmaticsEvaluation methods of anti-asthmatics
Evaluation methods of anti-asthmaticsNovo Nordisk India
 
Screening of anti alzheimers
Screening of anti alzheimersScreening of anti alzheimers
Screening of anti alzheimersDr Roohana Hasan
 
Screening of Antihyperlipidemic drugs
Screening of Antihyperlipidemic drugsScreening of Antihyperlipidemic drugs
Screening of Antihyperlipidemic drugsAbu Sufiyan Chhipa
 
Screening method of nootropics vikas malik
Screening method of nootropics vikas malikScreening method of nootropics vikas malik
Screening method of nootropics vikas malikVikasMalik68
 
pre clinical Screening for anti asthmatic drugs
pre clinical Screening  for anti asthmatic drugspre clinical Screening  for anti asthmatic drugs
pre clinical Screening for anti asthmatic drugsDHINESHKUMAR V
 
Pre clinical screening models for drugs acting on Autonomic nervous system
Pre clinical screening models for drugs acting on Autonomic nervous systemPre clinical screening models for drugs acting on Autonomic nervous system
Pre clinical screening models for drugs acting on Autonomic nervous systemRana Rana
 
Pharmacological screening by harikesh maurya
Pharmacological screening by harikesh mauryaPharmacological screening by harikesh maurya
Pharmacological screening by harikesh mauryaHarikesh Maurya
 
Screening models for immunomodulator
Screening models for immunomodulatorScreening models for immunomodulator
Screening models for immunomodulatorMr. MOHD FAHAD
 
In- vitro and in-vivo screening of Hypoglycemics.
In- vitro and in-vivo screening of Hypoglycemics.In- vitro and in-vivo screening of Hypoglycemics.
In- vitro and in-vivo screening of Hypoglycemics.Student
 
Screening of ANTIFERTILITY AGENTS and APHRODISIACS
Screening of ANTIFERTILITY AGENTS and APHRODISIACSScreening of ANTIFERTILITY AGENTS and APHRODISIACS
Screening of ANTIFERTILITY AGENTS and APHRODISIACSHimaniTailor
 
Screening of hepatoprotective drugs
Screening of hepatoprotective drugsScreening of hepatoprotective drugs
Screening of hepatoprotective drugsDipanjaliKamthe
 
screening of antiulcer agents
screening  of antiulcer agentsscreening  of antiulcer agents
screening of antiulcer agentsUttara Joshi
 

What's hot (20)

Screening models on aphrodisiac agents
Screening models on aphrodisiac agentsScreening models on aphrodisiac agents
Screening models on aphrodisiac agents
 
Hepatoprotective screening methods
Hepatoprotective screening methodsHepatoprotective screening methods
Hepatoprotective screening methods
 
screening of anti-fertility agent
screening of anti-fertility agentscreening of anti-fertility agent
screening of anti-fertility agent
 
Screening of antipyretic drugs
Screening of antipyretic drugsScreening of antipyretic drugs
Screening of antipyretic drugs
 
Screening models for aphrodisiac agents and anti fertility agents
Screening models for aphrodisiac agents and anti fertility agentsScreening models for aphrodisiac agents and anti fertility agents
Screening models for aphrodisiac agents and anti fertility agents
 
Screening of Diuretics M.PHARM PHARMACOLOGY.
Screening of Diuretics M.PHARM PHARMACOLOGY.Screening of Diuretics M.PHARM PHARMACOLOGY.
Screening of Diuretics M.PHARM PHARMACOLOGY.
 
Preclinical screening of anti fertility agents
Preclinical screening of anti fertility agentsPreclinical screening of anti fertility agents
Preclinical screening of anti fertility agents
 
Evaluation methods of anti-asthmatics
Evaluation methods of anti-asthmaticsEvaluation methods of anti-asthmatics
Evaluation methods of anti-asthmatics
 
Screening of anti alzheimers
Screening of anti alzheimersScreening of anti alzheimers
Screening of anti alzheimers
 
Screening of Antihyperlipidemic drugs
Screening of Antihyperlipidemic drugsScreening of Antihyperlipidemic drugs
Screening of Antihyperlipidemic drugs
 
Screening of antiparkinsonian agents
Screening of antiparkinsonian agentsScreening of antiparkinsonian agents
Screening of antiparkinsonian agents
 
Screening method of nootropics vikas malik
Screening method of nootropics vikas malikScreening method of nootropics vikas malik
Screening method of nootropics vikas malik
 
pre clinical Screening for anti asthmatic drugs
pre clinical Screening  for anti asthmatic drugspre clinical Screening  for anti asthmatic drugs
pre clinical Screening for anti asthmatic drugs
 
Pre clinical screening models for drugs acting on Autonomic nervous system
Pre clinical screening models for drugs acting on Autonomic nervous systemPre clinical screening models for drugs acting on Autonomic nervous system
Pre clinical screening models for drugs acting on Autonomic nervous system
 
Pharmacological screening by harikesh maurya
Pharmacological screening by harikesh mauryaPharmacological screening by harikesh maurya
Pharmacological screening by harikesh maurya
 
Screening models for immunomodulator
Screening models for immunomodulatorScreening models for immunomodulator
Screening models for immunomodulator
 
In- vitro and in-vivo screening of Hypoglycemics.
In- vitro and in-vivo screening of Hypoglycemics.In- vitro and in-vivo screening of Hypoglycemics.
In- vitro and in-vivo screening of Hypoglycemics.
 
Screening of ANTIFERTILITY AGENTS and APHRODISIACS
Screening of ANTIFERTILITY AGENTS and APHRODISIACSScreening of ANTIFERTILITY AGENTS and APHRODISIACS
Screening of ANTIFERTILITY AGENTS and APHRODISIACS
 
Screening of hepatoprotective drugs
Screening of hepatoprotective drugsScreening of hepatoprotective drugs
Screening of hepatoprotective drugs
 
screening of antiulcer agents
screening  of antiulcer agentsscreening  of antiulcer agents
screening of antiulcer agents
 

Similar to Evaluation of anticancer agents final

Anti- Tumor assay / Screening of Anticancer Drugs
Anti- Tumor assay / Screening of Anticancer DrugsAnti- Tumor assay / Screening of Anticancer Drugs
Anti- Tumor assay / Screening of Anticancer DrugsPratik Parikh
 
Preclinical Pharmacological Experiments.pptx
Preclinical Pharmacological Experiments.pptxPreclinical Pharmacological Experiments.pptx
Preclinical Pharmacological Experiments.pptxAkashKale71
 
new drug development by harsha
new drug development by harshanew drug development by harsha
new drug development by harshaSriharsha Rayam
 
Acute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animalsAcute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animalsSwaroopaNallabariki
 
Assignment on Toxicokinetics
Assignment on ToxicokineticsAssignment on Toxicokinetics
Assignment on ToxicokineticsDeepak Kumar
 
A review on stages of drug development and alternative methods for animal stu...
A review on stages of drug development and alternative methods for animal stu...A review on stages of drug development and alternative methods for animal stu...
A review on stages of drug development and alternative methods for animal stu...Frinto Francis
 
animal toxicity studies.pptx
animal toxicity studies.pptxanimal toxicity studies.pptx
animal toxicity studies.pptxDrRenuYadav2
 
FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...
FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...
FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...MedicReS
 
Pharmacological screening2 &amp; tox. studies
Pharmacological screening2 &amp; tox. studiesPharmacological screening2 &amp; tox. studies
Pharmacological screening2 &amp; tox. studiesNoor Alam
 
Screening of antimalarial drugs
Screening of antimalarial drugsScreening of antimalarial drugs
Screening of antimalarial drugsAnupam Prahlad
 
In-Vivo Safety - Pre Ind Drug Development
In-Vivo Safety - Pre Ind Drug DevelopmentIn-Vivo Safety - Pre Ind Drug Development
In-Vivo Safety - Pre Ind Drug DevelopmentOSUCCC - James
 
Alternatives to animal testing
Alternatives to animal testingAlternatives to animal testing
Alternatives to animal testingGandla Sowmya
 
Principles of cell viability assays by surendra.pptx
Principles of cell viability assays by surendra.pptxPrinciples of cell viability assays by surendra.pptx
Principles of cell viability assays by surendra.pptxSurendra Chowdary
 
Preclinical drug discovery and development
Preclinical drug discovery and developmentPreclinical drug discovery and development
Preclinical drug discovery and developmentsamthamby79
 
Preclinical trial for Anticancer drugs
Preclinical trial for Anticancer drugsPreclinical trial for Anticancer drugs
Preclinical trial for Anticancer drugsSanaspriya01
 
Alternatives to animal experiments
Alternatives to animal experimentsAlternatives to animal experiments
Alternatives to animal experimentsRoopali Somani
 
In vivo Carcinogenesity study PHARMACY.pptx
In vivo Carcinogenesity study PHARMACY.pptxIn vivo Carcinogenesity study PHARMACY.pptx
In vivo Carcinogenesity study PHARMACY.pptxsubhamsourajit1
 
7 viablity assay
7 viablity assay7 viablity assay
7 viablity assaybire12345
 

Similar to Evaluation of anticancer agents final (20)

Anti- Tumor assay / Screening of Anticancer Drugs
Anti- Tumor assay / Screening of Anticancer DrugsAnti- Tumor assay / Screening of Anticancer Drugs
Anti- Tumor assay / Screening of Anticancer Drugs
 
Preclinical Pharmacological Experiments.pptx
Preclinical Pharmacological Experiments.pptxPreclinical Pharmacological Experiments.pptx
Preclinical Pharmacological Experiments.pptx
 
new drug development by harsha
new drug development by harshanew drug development by harsha
new drug development by harsha
 
Acute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animalsAcute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animals
 
Assignment on Toxicokinetics
Assignment on ToxicokineticsAssignment on Toxicokinetics
Assignment on Toxicokinetics
 
A review on stages of drug development and alternative methods for animal stu...
A review on stages of drug development and alternative methods for animal stu...A review on stages of drug development and alternative methods for animal stu...
A review on stages of drug development and alternative methods for animal stu...
 
Muhamed.k
Muhamed.kMuhamed.k
Muhamed.k
 
animal toxicity studies.pptx
animal toxicity studies.pptxanimal toxicity studies.pptx
animal toxicity studies.pptx
 
Genotoxicity studies
Genotoxicity studiesGenotoxicity studies
Genotoxicity studies
 
FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...
FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...
FDA 2013 Clinical Investigator Training Course: Pharmacology/Toxicology in th...
 
Pharmacological screening2 &amp; tox. studies
Pharmacological screening2 &amp; tox. studiesPharmacological screening2 &amp; tox. studies
Pharmacological screening2 &amp; tox. studies
 
Screening of antimalarial drugs
Screening of antimalarial drugsScreening of antimalarial drugs
Screening of antimalarial drugs
 
In-Vivo Safety - Pre Ind Drug Development
In-Vivo Safety - Pre Ind Drug DevelopmentIn-Vivo Safety - Pre Ind Drug Development
In-Vivo Safety - Pre Ind Drug Development
 
Alternatives to animal testing
Alternatives to animal testingAlternatives to animal testing
Alternatives to animal testing
 
Principles of cell viability assays by surendra.pptx
Principles of cell viability assays by surendra.pptxPrinciples of cell viability assays by surendra.pptx
Principles of cell viability assays by surendra.pptx
 
Preclinical drug discovery and development
Preclinical drug discovery and developmentPreclinical drug discovery and development
Preclinical drug discovery and development
 
Preclinical trial for Anticancer drugs
Preclinical trial for Anticancer drugsPreclinical trial for Anticancer drugs
Preclinical trial for Anticancer drugs
 
Alternatives to animal experiments
Alternatives to animal experimentsAlternatives to animal experiments
Alternatives to animal experiments
 
In vivo Carcinogenesity study PHARMACY.pptx
In vivo Carcinogenesity study PHARMACY.pptxIn vivo Carcinogenesity study PHARMACY.pptx
In vivo Carcinogenesity study PHARMACY.pptx
 
7 viablity assay
7 viablity assay7 viablity assay
7 viablity assay
 

Evaluation of anticancer agents final

  • 1. Evaluation of anticancer agents - Dr Anup Thorat - Guide : Dr Sandhya Kamat
  • 2. History 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 2
  • 3. History 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 3 Louise Goodman Sidney Farber Father of modern chemotherapy Alfred Gilman
  • 4. Need for Novel Anticancer Agents • Multidrug resistance. • Long-term treatment with cancer drugs a/w severe side effects. • Cytotoxic drugs have the potential to be very harmful to the body unless they are very specific to cancer cells. • New drugs - more selective for tumour cells. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 4
  • 5. Pre clinical evaluation 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 5 IN VITRO IN VIVO
  • 6. In vitro cytotoxicity studies: • 1990 : NCI-60 screen • Cytotoxicity assays on panel of human cancer cell lines – MTT-assay – SRB- assay – 3H-thymidine uptake assay – Fluorescence Dye exclusion tests – Clonogenic assays – Cell counting assay 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 6
  • 7. In vitro cytotoxicity studies: Advantages • Reduce the usage of animals. • Less time consuming, • Cost effective & • Easy to manage • Able to process a larger number of compounds quickly with minimum quantity. • Range of concentrations used are comparable to that expected for in vivo studies Disadvantages: • Difficulty in maintaining of cultures. • Show negative results for the compounds which gets activated after body metabolism and vice versa. • Impossible to ascertain the Pharmacokinetics . 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 7
  • 8. Cell lines • Tumor cell lines derived from several cancer types – lung, colon, melanoma, renal, ovarian, brain, and leukemia. • The lines are prepared and cryopreserved using DMSO(di methyl sulfoxide ) • THAWING OF THE CELLS : Rapid thawing of frozen ampoule in water bath until it gets liquefied. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 8
  • 9. Cell lines • The solution is centrifuged with saline for 10 mins to remove the DMSO • The saline is discarded and aliquot is taken for cell counting, cell viability and for sub culturing. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 9
  • 10. CELL PROPERTIES & ASSAY PROPERTIES ASSAY ENZYMATIC PROPERTY MTT ASSAY MEMBRANE INTEGRITY DYE EXCLUSION TEST DNA CONTENT/REPLICATION STATUS 3H-THYMIDINE UPTAKE & FLUORESCENT ASSAY PROTEIN CONTENT SRB ASSAY COLONY FORMING POTENTIAL CLONOGENIC ASSAY CELL DIVISION CELL COUNTING ASSAY 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 10
  • 11. Microculture Tetrazolium Test(MTT Assay) • A quantitative colorimetric assay • Measures cellular growth, cell survival and cell proliferation based on the ability of living cells. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 11 MTT (3-(4,5- Dimethylthiazol-2-yl)- 2,5- diphenyltetrazolium bromide) Formazan mitochondrial dehydrogenase in living cells only
  • 12. MTT Assay - Procedure • Cells from particular cell lines in log phase of growth are trypsinised. Check the cell viability through haemocytometer . • Adjust to appropriate density in suitable medium and inoculate in multiwell plates. • Cells are treated with various conc. of test compounds & the plate incubated at 37 °C in 5% CO2 /95% humidified air (1-4 d) • Cultures are taken out and 10 μl of MTT dye is added (5 mg/ml) into each well and incubated for 4hrs 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 12
  • 13. MTT Assay 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 13
  • 14. MTT Assay - Procedure • The plate is centrifuged, supernatant discarded & precipitated formazan salt is dissolved in 100 μl of isopropanol/DMSO • The plate samples are read at 570 nm microtiter plate reader IC50 of drugs can be determined by counting the viable cells % Cell viability = Absorbance of treated cells x 100 (%MTT reduction) Absorbance of untreated cells 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 14
  • 15. Microtiter plate reader 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 15
  • 16. SRB (SULPHORHADAMINE B) ASSAY • Measures whole protein content which is proportional to the cell number. • SRB – a bright pink anionic protein staining dye that binds to the basic amino acids of the cellular proteins. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 16
  • 17. SRB ASSAY - Procedure • Cell lines are counted, cultured & inoculated in 96 well plates. • After incubation with different concentrations of test compounds, the cell cultures are stained with SRB dye. • Washing with acetic acid removes the unbound dye and the protein bounded dye is extracted using triss base • Optical density is determined by 96-well plate reader 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 17
  • 18. 3H-Thymidine uptake assay • Provides information about tumour kinetics, ploidy status of the cells. Procedure: 1. Tumor cell suspensions are exposed to the drug for 5 days. 2. Radio-labeled 3H-thymidine is added 3. Replicating cells will incorporate 3H-thymidine into their DNA 4. Determined by autoradiography or liquid scintillation. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 18
  • 19. FLUORESCENCE ASSAY • Cells are exposed to fluorescent labeled precursors after drug exposure. • Replicating cells will incorporate the dye into their DNA. • Resulting fluorescence is measured by flow cytometry / fluorescent microscopy . 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 19
  • 20. Dye Exclusion Test • This assay is relayed on the structural integrity of the cells. • Dyes used : Trypan blue, Eosin, or Nigrosin • Live cells possess intact cell membranes that exclude the dye, whereas dead cells having lost membrane integrity take up the dyes. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 20
  • 21. Dye Exclusion Test - Procedure • Cells are incubated with different concentrations of test compounds for 4 days. • Dead cells are stained with dye colour. • Specimen is centrifuged & collected in microscopic slides. • Live cells – stained with hematoxylin eosin. • Tumor cell cytotoxicity is compared with control – duck erythrocytes. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 21
  • 22. Trypan blue dye exclusion test Live cell Dead cell 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 22
  • 23. CLONOGENIC ASSAY • Measures the growth inhibition. • Important for the drugs that act by arresting the cells at checkpoints in the cell cycle Procedure: 1. Single cell suspensions are exposed to anticancer agents to be tested. 2. Suspensions are rinsed and plated in a semisolid medium. 3. After 14 to 28 days, some cells will having undergone several division form tumour colonies which can be quantified in a visual or semi- automated fashion. 4. No of colonies from treated cells is compared with untreated colonies. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 23
  • 24. CLONOGENIC ASSAY 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 24
  • 25.
  • 26. Preclinical Toxicity Studies • Aimed at predicting (a) Safe starting dose & dosage regimen for human clinical trials(P1) (b) The toxicities of the compound, & (c) The likely severity and reversibility of drug toxicities. • Regulatory requirement : Two acute preclinical toxicity studies 1. Rodent (mice) - single- and multiple-dose lethality studies. 2. Non rodent (dogs) - single- and multiple-dose confirmatory toxicity. • Cytotoxic & non cyotoxic drugs 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 26
  • 27. Acute Toxicity Studies • First mouse given a single injection (IP, IV, SC, IM or PO) of 400 mg/kg (or lower if the compound is extremely potent) • Second mouse - 200 mg/kg & • Third mouse - 100 mg/kg • The mice are observed for a period of 2 weeks & sacrificed if there are signs of significant toxicity • If all 3 mice must be sacrificed, the next 3 dose levels (50, 35 and 12.5 mg/kg) are tested in a similar manner • This process is repeated until a tolerated dose is found designated as MTD. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 27
  • 28. Preclinical Toxicity Studies • To determine the phase I entry dose of a cytotoxic anticancer agent, the dose levels that are lethal to 10, 50, and 90% of mice (LD10, LD50, & LD90) is determined by the same route of administration. The projected phase I entry dose is usually 1/10th of the LD10 • A robust toxicity study needed to avoid lengthy phase I trials as well as severe drug toxicities. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 28
  • 29. Why Mice? • Mice are small, easy to handle. • Short generation time & accelerated lifespan,manageable costs, space, and time. • Striking similarity to humans in anatomy, physiology, and genetics. • Over 95% of the mouse genome is similar to humans. • Many of the genes responsible for complex diseases are shared between mice and humans. • Mouse genome can be directly manipulated. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 29
  • 30. Tumour Models 1. Carcinogen induced models 2. Viral infection models 3. Transplantation Models – Murine – Human 4. Genetically Engineered Mouse Models 5. In vivo hollow fibre assay 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 30
  • 31. CHEMICAL CARCINOGEN MODEL DMBA induced mouse skin papillomas • Two stage experimental carcinogenesis – Initiator – DMBA (dimethylbenz[a]anthracene), – Promotor – TPA. (12-O-tetradecanoyl-phorbol-13-acetate) • Mice : Single dose – 2.5 µg of DMBA f/b 5 to 10 μg of TPA in 0.2 ml of acetone twice weekly. • Papilloma begins to appear after 8 to 10 wks - Tumor incidence & multiplicity of treatment group is compared with DMBA control group 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 31
  • 32. Mouse skin papillomas 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 32
  • 33. MNU INDUCED RAT MAMMARY GLAND CA • Induces hormone dependent tumors. • Single i.v of 50 mg/kg of methylnitrosourea - 50 days old SD rats. • Adenocarcinoma will be produced within 180 days of post – carcinogen – 75 to 95% • Reduction in tumor size is compared • Drawback – cannot detect inhibition of carcinogen activation. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 33
  • 34. RAT MAMMARY GLAND CA 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 34
  • 35. DMBA induced rat mammary gland ca • Sprague dawley rats • Intragastric injection of 12 mg / kg of DMBA at 50 days of age • Cancer appears within 120 days – 80 to 100% • Detect the drugs inhibiting carcinogen activation. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 35
  • 36. MNU induced tracheal squamous cell ca • Hamster • 5% of MNU in normal saline is given once a week for 15 wks by catheter. • Within 6 months – 40 to 50% • Test drug efficacy is measured by comparing with carcinogen control group 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 36
  • 37. DEN induced lung adenocarcinoma in Hamster • 17.8mg DEN/kg body wt twice weekly by S.C inj for 20 weeks starting at age 7 to 8 weeks. • Usually produces tracheal tumors in 90-100% and lung tumors in 40-50% of male syrian hamster. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 37
  • 38. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 38 Cancer site Cancer Type Species Carcinogen Colon Adenocarcinomas Rat AOM (azoxymethane) Prostate Adenocarcinomas Rat MNU (methylnitrosourea) Esophagus Squamous cell carcinoma Rat NMBA(N-nitroso- methylbenzylamine) Breast Adenocarcinoma Mice NMU (N-Nitroso-N- methylurea)
  • 39. Viral infection models • Mouse Mammary Tumor Virus (MMTV) was the first mouse virus, isolated at Jackson labs as the “non-chromosomal factor” that caused mammary tumors in the C3H strain of mice. • Some viruses cause cancer via random integration in certain cells • Some viruses carry cellular oncogenes – Abelson murine leukemia virus – Abl – Moloneymurine sarcoma virus – Raf • Engineered viruses now used routinely in the laboratory to induce cancer. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 39
  • 40. Transplantation Models • Tumor cells or tissues (mouse or human) transplanted into a host mouse. • Ectopic – Implanted into a different organ than the original (typically subcutaneous or kidney capsule) • Orthotopic – Implanted into the analogous organ of the original tumor. • Advantages : – Typically cheap, fast & easy to use. – Not covered by patents 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 40
  • 41. Transplantation Models • Disadvantages: – Histopathology cannot be exactly correlated to human tumors – Lack of step-wise progression through pre-neoplastic stages – Evolution of tumor cells during passaging – Requirement for angiogenesis to support the newly transplanted tumor 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 41
  • 42. Transplantation Models : Human Tumor Xenografts • Athymic “nude”mice developed in 1960’s • Mutation in nu gene on chromosome 11 • Phenotype: retarded growth, low fertility, no fur, immunocompromised – Lack thymus gland, T-cell immunity • First human tumor xenograft of colon adenocarcinoma by Rygaard & Poulson, 1969 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 42
  • 43. Xenograft Sites • Subcutaneous tumor (NCI method of choice) with IP drug administration • Intraperitoneal • Intracranial • Intrasplenic • Renal subcapsule • Site-specific (orthotopic) organ inoculation 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 43
  • 44. Xenograft Study Endpoints • Toxicity Endpoints: – Drug related death – Net animal weight loss • Efficacy Endpoints: – Tumor weight change – Treated/control survival ratio – Tumor growth assay (corrected for tumor doubling time) 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 44
  • 45. Xenograft Tumor Weight Change • Tumor weight change ratio (used by the NCI in xenograft evaluation) • Defined as: treated/control x 100% • Tumor weight in mg = (a x b2)/2 – a = tumor length – b = tumor width • T/C < 40-50% is considered significant 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 45
  • 46. Human Tumor Xenografts Advantages • Many different human tumor cell lines transplantable • Wide representation of most human solid tumors • Good correlation with drug regimens active in human lung, colon, breast, and melanoma cancers Disadvantages • Brain tumors difficult to model • Different biological behavior, metastases rare – Survival not an ideal endpoint: death from bulk of tumor, not invasion • Shorter doubling times than original growth in human • Difficult to maintain animals due to infection risks 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 46
  • 47. Transplantation Models : Cell line based allografts 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 47
  • 48. Transplantation Models : Cell line based allografts Advantages • Very easy to maintain cell lines indefinitely; • Very fast to generate large numbers of tumors • Relative homogeneity of tumors makes it easy to detect differences • Comparatively cheap • Intact immune system Disadvantages • Mouse cells rather than human • Inaccurate histopathology compared to human tumors • Evolution of tumor cells during culture • As it involves murine CA, ability to predict response to therapy in humans is controversial. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 48
  • 49. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 49
  • 50. In Vivo Hollow Fibre Assay • In vivo screening tool implemented in 1995 by NCI • 12 human tumor cell lines (lung, breast, colon, melanoma, ovary, and glioma • Cells suspended into hollow polyvinylidene fluoride fibers implanted IP or SC in lab mice • After in vivo drug treatment, fibers are removed and analyzed in vitro • Antitumor (growth inhibitory) activity assessed 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 50
  • 51. In Vivo Hollow Fibre Assay 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 51
  • 52. In Vivo Hollow Fibre Assay Subcutaneous Hollow Fibre implants 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 52
  • 54. Clinical Evaluation • The most reliable method for demonstrating efficacy is to show a statistically significant improvement in a clinically meaningful endpoint in blinded randomized controlled trials 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 54
  • 55. Regulatory considerations 1. Regular Approval – Longstanding route of drug approval based on the demonstration of clinical benefit 2. Accelerated Approval – Use of a surrogate endpoint that is reasonably likely to predict benefit. – Serious or life-threatening diseases – Improvement over available therapy – No existing therapy 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 55
  • 56. Endpoints • 1970s - Objective Response Rate (ORR) • 1980s - improvement in survival, quality of life (QOL), physical functioning, tumour related symptoms. • Three kinds of end points have been used: 1. Overall Survival 2. Tumor assessment endpoints e.g. ORR,DFS, PFS,TTP, TTF; and 3. Symptom assessment endpoints e.g. palliation of side effects, QOL scores. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 56
  • 57. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 57 Endpoint Regulatory Evidence Study Design Advantages Disadvantages Overall Survival (OS) Clinical benefit for regular approval Randomized studies essential Blinding not essential 1)Universally accepted direct measure of benefit 2)Easily & Precisely measured 1)May involve larger studies 2)May be affected by crossover therapy and sequential therapy 3)Includes non cancer deaths Symptom Endpoints (patient reported Outcomes) Clinical benefit for regular approval Randomized blinded studies 1)Patient perspective of direct clinical benefit 1)Blinding is often difficult 2)Data are frequently missing or incomplete 3)Clinical significance of small changes is unknown 4)Multiple analyses 5)Lack of validated instruments Objective Response Rate (ORR) Surrogate for accelerated Approval or regular Single-arm or randomized studies can be used Blinding 1)Can be assessed in single-arm studies 2)Assessed earlier and in smaller studies compared with survival 1)Not a direct measure of benefit 2)Not a comprehensive measure of drug activity 3)Only a subset of patients
  • 59. Single-Arm Studies • No available therapy • ORR (objective response rate) and response duration in single-arm studies can be a substantial evidence supporting accelerated approval. • Primary efficacy measure : Proportion of patients who achieve a complete or partial response to the treatment. • Design eliminates truly ineffective therapy 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 59
  • 60. Single-Arm Studies • Challenges : 1. Do not adequately characterize time-to-event endpoints (survival, TTP, PFS) 2. Because of variability in the natural history of cancer, a randomized study is necessary to evaluate time-to-event endpoints. 3. Inability to predict comparative performance vis-a`-vis the then- available best possible, standard-of-care therapeutic option. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 60
  • 61. Non-inferiority Studies • Rely on historical data to establish the active control’s treatment effect size & constancy assumption • Estimated size of the active-control’s treatment effect should be based on a comprehensive meta-analysis of historical studies • Challenges : 1. the estimation of active-control effect and the determination of amount of effect (NI margin) to be retained. Usually large sample sizes. 2. Subsequent therapies and crossover to the active-control arm can confound any NI analysis 3. NI trials with endpoints other than survival are problematic 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 61
  • 62. Clinical Trial Phases • What is the dose and schedule? Phase I • Is it active? – spectrum of anticancer activity ? Phase II – is it better than standard therapy? Phase III • Is it safe? Phase I to IV 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 62
  • 63. Phase 0 : Microdosing • The FIH dose : significant safety risk for the patient volunteers. • Less than1/100th of the dose calculated to yield a pharmacological effect of the test substance to a maximum dose of less than 100 µg. • Advantages: – Mitigate FIH dose risk, – Gather early pharmacokinetic data (bioavailability, clearance, elimination rate), and – Increased efficiency of drug development. • Highly sensitive analytical methods : PET, LCMS, accelerated mass spectroscopy(AMS) 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 63
  • 64. Phase 1 • Usually conducted in patients, rather than healthy volunteers. • This adds to the challenges : 1. Recruitment of tumor-specific patient volunteers 2. The recruited volunteers may be in the advanced stages of the disease - refractory to the currently available standard-of-care treatment options - cost escalation. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 64
  • 65. Dose Escalation Studies • Phase 1 entry dose is obtained from preclinical toxicity studies • Endpoints : – Along with MTD, info on clinical toxicity, pharmacokinetics, and preliminary antitumor activity is obtained. – Also for cytotoxic agents, dose-related toxicity is regarded as a surrogate for efficacy. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 65 Progressively escalating doses MTD
  • 66. Dose Escalation Studies • Modified Fibonacci search : 1. Safe starting dose(d) = 1/10th of the LD10 2. Enroll patients in cohorts of three, and escalate the dose according to a modified Fibonacci sequence 3. Higher escalation steps have decreasing relative increments -100, 65, 52, 40, 29% & thereafter 33% increases over the previous dose. 4. Ethical issues : substantial numbers of patients are treated at nontherapeutic doses 5. Efficiency issues : Lengthy trials, time & cost incurred. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 66
  • 67. Dose escalation : Other Methods • Pharmacologically guided dose escalation (PGDE) – Using the preclinical toxicology data to rapidly escalate doses to a target area under the curve (AUC) value obtained from murine pharmacokinetic data. • Non-pharmacokinetic statistical modeling approaches : – Statistical approaches model the dose–toxicity relationship as a sigmoidal curve to predict the MTD. The toxicity is then evaluated & actual MTD found by rapid dose titration. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 67
  • 68. Ethical Challenges In Phase I 1. The paucity of benefits with substantial risks 2. Informed consent • Majority of participants are treated at doses that cannot produce responses in human tumors. • The objections based on informed consent are deficiencies of disclosure, understanding, and voluntariness • Vulnerable group – Terminally ill patient 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 68
  • 69. Phase II and III Clinical Trials • Phase II studies are carried out in a small group of patients with a specific tumor type to – Determine anticancer efficacy & – To define the therapeutic window of the compound • Any arm may be terminated early because of discouraging results, and the response rate of each arm is assessed separately against a historical control rate with definable α and β error probabilities • Phase III trials are conducted in a much greater number of patient volunteers of the selected tumor type with prospective and randomized evaluation against standard of care. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 69
  • 70. Phase II and III Clinical Trials • Embedding a randomized phase II study within a phase III study – Patients who progress on one of the arms of Phase II would then be randomly assigned to arms E or C of phase III & and – Their survival data would be combined in a stratified fashion with data from patients directly assigned to arms C or E for the phase III assessment. – This design allows untreated patients to be enrolled onto the phase II study, while assuring them a more established therapy if they progress 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 70
  • 71. Phase II and III Clinical Trials • Phase II studies act as a screen of antitumor efficacy to select the most promising agents to enter the pivotal phase III clinical trials. • A Phase II study should efficiently eliminate truly ineffective therapy & reliably indicate whether subsequent phase III testing is warranted. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 71
  • 72. 5/4/2014 Evaluation of anticancer agents - Dr. Anup Thorat 72

Editor's Notes

  1. Chemical warfare.
  2. G & G story. Farber story - Mtx : remission in ALL
  3. MDR : Blood , breast, ovarian, lung, lower GIT
  4. NCI60 screen generates D-R curves of potential compounds representing diff profiles of molecular markers & biochemical pathways. This info on cell inhibition is used to generate Fingerprint of the compound. Comparing fingerprint with lib database of compounds with known MOAs helps hypothesize the MOA of novel compounds.
  5. DMSO(di methyl sulfoxide ) which preserve the cell during freezing. DMSO is toxic at room temperature.
  6. Quantifying the mitochondrial metabolism and respiratory chain activity of cells Yellow MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) a tetrazolium salt is reduced to purple formazan by mitochondrial dehydrogenase of living cells in which tetrazolium ring gets cleaved in mitochondria
  7. Starting 20µg/ml
  8. causing the live tissues to turn deep purple in color. Test materials that result in cell death will not produce this color change. The more toxic the test material, the less purple the tissue will be.
  9. Colorimetric assay for the quantification of the total protein synthesis rate of cells
  10. DNA CONTENT/REPLICATION STATUS
  11. Colony forming potential
  12. For anticancer drugs their efficacy is closely related to their toxicity. Bcoz their therapeutic effect and toxicity are attributed to the same mechanism. Pre clinical toxicity data is important for regulatory approval. Cytotoxic-acute 28days ; noncytotoxic – rodent 6months, non rodent 12 months
  13. If serious, irreversible toxicities are exhibited in the non-rodent species at the projected starting dose, then the human starting dose is reduced to the 1/6thof the highest dose tested. Overestimation-toxicity-fludarabine-marrow suppression. Underestimation-cost, time, unethical low dose
  14. Dogs, cats, horses, pigs, and goats. baboons, chimpanzees, macaques, marmosets and tamarins Accelerated lifespan (one mouse year equals about 30 human years),
  15. MNU, methylnitrosourea
  16. DEN : diethylnitrosamine
  17. Summarise
  18. Patient-derived xenografts
  19. Useful in targeted therapy
  20. Condition: Manufacturer conducts further studies to verify actual clinical benefit including postmarketing studies. FDA recommends at least two adequate and well-controlled clinical trials.
  21. 1980s- should be based on more direct evidence of clinical benefit. Tumour assessment end point selection consider 2 judgements. First, a determination of whether the endpoint will support either accelerated approval or regular approval should be ascertained. Second, the endpoint should be evaluated for the potential of bias or uncertainty in tumor endpoint assessments.
  22. OS : the time from randomization to time of death from any cause. ORR is the proportion of patients with tumor size reduction of a predefined amount and for a minimum time period. Not useful for stable disease DFS is the time from randomization until recurrence of tumor or death from any cause. *Adjuvant setting.*DFS can be an important endpoint in situations where survival may be prolonged, making a survival endpoint impractical. PFS is the time from randomization to objective tumor progression or death. TTP is defined as the time from randomization until objective tumor progression. Does not include deaths
  23. Utilizing historical controls on the currently best-available treatment. Acute leukemia: complete responses a/w decreased transfusion requirements, decrease in infections, and increased survival
  24. Constancy assumption- control grp effect has remained constant between the historical study and the current study. Assumes constancy of patient population characteristics, supportive care measures, and evaluation techniques .
  25. Concept is based on using extremely low doses of a drug, which are pharmacologically inactive but are able to delineate the pharmacokinetic profile of the drug in humans. Drug with nonlinear pharmacokinetics - inability to predict PK parameters.
  26. identify the MTD