Early Clinical Cancer Trials Phase I and II studies - PowerPoint PPT Presentation

1 / 37
About This Presentation
Title:

Early Clinical Cancer Trials Phase I and II studies

Description:

Pre-clinical proof of principle must be achieved ... Pre-clinical antitumor activity. ethics (age, life-expectancy, PS, risks, follow-up) ... – PowerPoint PPT presentation

Number of Views:172
Avg rating:3.0/5.0
Slides: 38
Provided by: ulrikl
Category:

less

Transcript and Presenter's Notes

Title: Early Clinical Cancer Trials Phase I and II studies


1
Early Clinical Cancer Trials Phase I and II
studies
  • Ulrik Lassen, MD., PH.D.
  • Head of Phase I Unit
  • Dept. Of Oncology, Finsen Center
  • Rigshospitalet, Copenhagen

2
Research in
  • New superior agents improving survival
  • New agents with fewer adverse events 
  • New agents against side effects
  • Reducing treatment periods
  • More efficacious radiotherapy
  • Methods to protect normal tissue during
    radiotherapy

3
Phases in drug development
  • Pre-clinical studies proof of concept
  • Phase 1 dose-finding
  • Phase 2 activity and safety
  • Phase 3 randomised trials

The whole process takes 6-12 years and costs 1
billion DKK for each approved agent
4
Industry loves oncology
  • Robert H. Glassman
  • Merryll Lynch, N.Y.

ENA, Philadelphia, 2006
5
Why?
  • 1/3 of all drugs in development are cancer drugs
  • Targatable physicians (few oncologists in
    relation to the number of studies)
  • Cancer drugs represent the largest income
  • 1300 drugs in late discovery in USA
  • 600 drugs in clinical evaluation
  • 95 approvals/year (FDA)
  • 3-5 approved/year (FDA)

6
But FDA is not quite satisfied
  • The rate of success for phase III studies have
    dropped from 50 to 30 in 1990-2004
  • The rate of success is higher for other
    indications- CNS 20- Oncology 30-
    CV 50- Others 70

7
How come?
  • Dropping response rates in phase II (low hanging
    fruits have been picked)
  • To poor target validation for new drugs- 1990
    gt100 references at clinical entre- 2000 only
    8-10 references
  • 200 inputs 2-3 approvals

8
Phase I - background
  • First in man studies
  • Pre-clinical proof of principle must be achieved
  • Chemical, pharmaceutical and animal
    pharmaco-logical and toxicological data must be
    available
  • Purpose to describe adverse events and
    pharmacokinetic and dynamic profiles
  • Identify organs at risk

9
Classic phase I design
  • Dose-escalation
  • 3-6 patients/dose level
  • MTD, DLT
  • Recommended doseToxicity as surrogate endpoint

10
Typical DLT definitionPatients experiencing one
of the following toxicities in cycle one are
considered as having dose-limiting toxicity (DLT)
(see NCI/CTC)
  • Absolute neutrofile count lt 0,5 x 109/l lasting
    ?7 days, or absolute neutrofilte count lt 0,5 x
    109/l with septicaemia
  • Absolute platelet count lt 25 x 109/l lasting ?7
    days, or platelets lt 10 x 109/l
  • Any other drug-related non-haematological grade
    3-4 toxicity, except alopecia, nausea and
    vomiting, skin-rash, artralgias and myalgias, if
    appropriate prophylactic action have not been
    taken.

11
What is CTCAE v3.0
  • Definitions and grading
  • General or organ specific
  • Acute, late or chronic
  • Symptomatic or asymptomatic
  • Subjective or objective (radiographic or
    biochemical)
  • Does not discriminate between cause or error
  • CTCAE v3.0 is developed as a tool for clinical,
    scientific studies, and not for routine use or
    standard therapy

12
Grading
  • 0 No adverse event or within normal values
  • 1 Mild adverse event
  • 2 Moderate adverse event
  • 3 Serious and undesired adverse event
  • 4 Life-threatening or invalidating adverse
    event
  • 5 Death related to adverse event

13
Classic phase I design - again
  • Dose-escalation
  • 3-6 patients/dose level
  • MTD, DLT
  • Recommended doseToxicity as surrogate endpoint

14
Definitions
10 of lethal dose in test animals, Expressed in
mg/m2
  • LD10

DLT
Dose limiting toxicity
incl. 3 more pts.
example
Toxicity in 1/3
No toxicity
dose-escalation
More toxicity
stop dose-escalation
stop dose-escalation
Toxicity in gt 1/3
Maximal tolerable dose (tox. lt 1/3)
MTD
15
Dose-escalation - fibonacci (modified)
  • first dose level (most often 1/10 of LD10)
  • second dose level twice first dose
  • third dose level 67 greater than the second
  • Fourth dose level 50 greater than the third
  • Fifth dose level 40 greater than the
    fourth
  • Sixth dose level 33 greater than the
    fifthNo dose escalation in the same patient

16
Dose-escalation, PK-guided
  • MTD - LD10 in mice
  • Linear PK necessary
  • Valid assays
  • Caution inter-patient variability
  • If necessary, wait for the results
  • Continuous reassessment method
  • Preferable at wide therapeutic ratios
  • 1 patient per dose level
  • Escalate 100 if no toxicity
  • If toxicity, then include more patients
  • Caution Sudden, unexpected DLT Dangerous?
    Cumulative toxicity

17
Phase I - patients
  • Healthy volunteers (not cytostatics)- high risk
    groups in chemoprevention
  • Cancer patients without other treatment options
    (therapy)
  • Life expectancy gt 12 weeks
  • Different tumor types
  • PS 0, 1 and possibly 2

18
Phase I biological agents
  • Antibodies, antisense-nucleotides, gene-therapy
  • Unknown therapeutic ratios
  • Unknown dose-response relations
  • DLT may never be reached
  • What is first dose level in case of no AE in
    animals?
  • Endpoints? pharmacodynamic, biomarkers-enzyme-in
    hibition, methylation, phosphorylation,
    acethylation, biopsies (skin, tumor), PET scans

19
Examples biological agents
  • Anti-EGF direct from phase I to phase III
  • Interferon anti-angiogenetic features seen at
    doses below MTD, but not at MTD
  • Gene therapy?
  • HDAC acetylation of histones in peripheral
    mononuclear blood cells
  • TKI phosphorylation of downstream targets
  • Apoptosis inhibitors grade of apoptosis

20
Other phase I trials
  • Phase Ib -Phase 1 trial in similar patients
    (one tumor type)-e.g. a distinct target
  • Phase I-II -One tumor type, more
    patients-Combinations of investigational agents
    and known cytostatics-Dose-escalation of more
    agents-Determine dose and sequence of
    agents-Endpoints safety and activity

21
Importance of phase 1 trials
  • Drug development
  • Responding tumor types of importance to phase 2
    trials
  • DLT, MTD and PK not only endpoints, but also
    indicators for the optimal dose for the desired
    target
  • Benefit-risk ratio high
  • Imatinib had 93 OR in phase 1 for CML

22
Limitations for phase 1 trials
  • Many patients get homeopatic doses below the
    biologic active level
  • Few patients get benefit (approx. 5-10 objective
    response, but up to 50 get stabilisation/symptom
    relief)
  • Time consuming trials (observation of kohorts
    prior to dose escalation)
  • Problems with inter-patient-variability
  • Toxicity as a surrogate marker cannot be used if
    agent is non-toxic

23
Response Rates According to Year
Horstmann, E. et al. N Engl J Med 2005352895-904
24
Consequenses
  • Phase 1 trials are becoming small because new
    targeted agents are less toxic
  • The challenge is now phase 2 design - how to
    interpret low response rate but long-lasting
    stabilisation (e.g. erlotinib, bevacizumab,
    cetuximab)
  • Can we drop phase 2?
  • No, not according to the experiences from MMI,
    FTI and gefitinib

Ratain and Eckhardt, JCO 22, 2004
25
Whats next?
  • Phase 2 must preventive negative phase 3 trials
  • Randomised phase 2 studies with cross-over,
    perhaps with more tumor types and separate
    analyses
  • Valid expectations for PR and SD
  • Time to progression as endpoint
  • Valid biomarkers

Ratain and Eckhardt, JCO 22, 2004
26
Phase II - therapeutic pilot studies
  • Early phase II antitumor activity toxicity
    pharmacokinetics
  • Late phase II Confirm early (more pt.)
    pharmacokinetics other study populations

27
Phase II - patients, tumor types
  • Based on the agent mode of action
  • Phase II activity (early phase II)
  • Pre-clinical antitumor activity
  • ethics (age, life-expectancy, PS, risks,
    follow-up)

28
Phase II - endpoints
  • Response rate as a surrogate, not necessarily
    related to change of symptoms
  • Time-intervals preferable time to
    progressionmedian survival and overall survival
  • Safety, toxicity
  • Quality of life, symptom relief
  • NB. comparisons to other studies not allowed
  • Phase II results always superior to phase III

29
Follow up - methods
  • Phase 1 Adverse events- CTCAE version 3.0
  • Phase 2 Response or other surrogate markers and
    adverse events- RECIST or WHO
  • Phase 3 survival, adverse events and QoL

30
Only patients with measurable disease at baseline
should be included in protocols where objective
tumor response is the primary endpoint
  • RECIST
  • Measurable disease - the presence of at least one
    measurable lesion. If the measurable disease is
    restricted to a solitary lesion, its neoplastic
    nature should be confirmed by cytology/histology.
  • Measurable lesions - lesions that can be
    accurately measured in at least one dimension
    with longest diameter ?20 mm using conventional
    techniques or ?10 mm with spiral CT scan.
  • Non-measurable lesions - all other lesions,
    including small lesions (longest diameter lt20 mm
    with conventional techniques or lt10 mm with
    spiral CT scan), i.e., bone lesions,
    leptomeningeal disease, ascites,
    pleural/pericardial effusion, inflammatory breast
    disease, lymphangitis cutis/pulmonis, cystic
    lesions, and also abdominal masses that are not
    confirmed and followed by imaging techniques

31
Evaluation of target lesions
  • Complete Response (CR) - Disappearance of
    all target lesions
  • Partial Response (PR) - At least a 30
    decrease in the sum of the LD of target lesions,
    taking as reference the baseline sum LD
  • Progressive Disease (PD) - At least a 20
    increase in the sum of the LD of target lesions,
    taking as reference the smallest sum LD recorded
    since the treatment started or the appearance
    of one or more new lesions
  • Stable Disease (SD) - Neither sufficient
    shrinkage to qualify for PR nor sufficient
    increase to qualify for PD, taking as reference
    the smallest sum LD since the treatment started

32
Standard phase II
  • Pro- equivocal, quick, cheap, well-defined
    endpoints/surrogate endpoints
  • Contra-selection bias for endpoints (response,
    progressions-free survival and survival)

33
Randomised phase II
  • Two or more parallel phase II studies
  • Equal patient populations
  • No comparison (too few patients)
  • No reference arm, only investigational arms
  • Indicator for which arm to enter phase III

34
Randomised phase II
  • Pro- better response estimates, duration and
    DFS- some protection against selection bias,
    better statistics to evaluate toxicity
  • Contra- poor power, time-consuming, expensive-
    endpoints not well accepted- cannot substitute
    phase III

Remember phase II for learning - phase III for
confirming Sheiner LB, Clin Pharmacol Ther
1997
35
Phase I Unit, Dept. of OncologyRigshospitalet,
Copenhagen
  • Dedicated unit for experimental cancer therapy
    and phase I trials.
  • Offer complete project management and clinical
    trial management systems.
  • Operating with ICH GCP to industry standards,
    including standard operating procedures (SOPs)
    covering all aspects of running clinical trials.
  • Comply with all current legal requirements and
    the needs of the EU Directive on Clinical Trials
    (Directive 2001/20/EC).
  • Part of a network of leading scientists and
    oncologists, including collaboration with other
    phase I units in Europe and the US.

36
Pharmacokinetics and pharmacodynamics
  • Detailed pharmacokinetic (PK) and pharmacodynamic
    (PD) analyses are key components of phase I and
    II clinical trials.
  • This includes biological and pharmacological
    studies on the new agent to ensure that it is
    acting by its proposed mechanism in patients and
    that a potentially active drug concentration can
    be achieved and maintained.
  • Our own staff of trained and GCP-examined
    research nurses obtain and handle blood samples
    for PK and PD in the laboratory facilities in the
    phase I unit.
  • Tissue sampling and processing for further
    analysis including snap-freeze technique can be
    undertaken through our collaboration with the
    department of diagnostic radiology and the
    surgical departments.

37
Immunological and clonogenic assays Through our
network of local academic laboratories at
Rigshospitalet, molecular biological/biochemical
analyses can be undertaken, including
  • Immunohistochemistry and other immunological
    assays
  • Micro-array gene analyses
  • Chromosomal analyses
  • Receptor expression and functional receptor
    analyses
  • Proteomics
  • Our clinic has a close collaboration with
    in-house laboratories for experimental basic
    research in oncology.
Write a Comment
User Comments (0)
About PowerShow.com