SlideShare una empresa de Scribd logo
1 de 10
Descargar para leer sin conexión
Make life simpler


Translational Pharmacology:
The Key to Ensuring Safe and Effective Dosing
L. Bruce Pearce, Ph.D1., Sue McKendrick, MSc, CStat2, John J. Jessop, Ph.D. 1, Julia Barrett,
M.D. 1 and Mark Baird, BSc(Hons)2
1
  Biologics Consulting Group, Inc. Alexandria VA, 2Quanticate, Ltd. Hitchin, UK


Introduction:

The relationship between dose, systemic exposure, and both safety and efficacy are the most
important elements in biopharmaceutical/drug development. The key is to attain adequate
systemic exposure to mediate a clinically effective response while avoiding a level of exposure
that results in toxicity. Understanding these relationships constitutes the central dogma of
product development from preclinical studies and on through to Phase 1, 2, and 3 clinical trials,
and is still important in post-marketing studies and pharmacovigilance.

In order to proceed successfully through clinical development, it is necessary to accurately
assess and demonstrate a favorable risk/benefit relationship at each milestone. This begins at
the preclinical level, where appropriate pharmacodynamic, safety pharmacology,
pharmacokinetic, toxicology and toxicokinetic studies are performed in various animal species.
The results of these studies are used to demonstrate proof-of-concept for the proposed
indication and to support safety for first-in-human administration in a Phase I clinical trial. These
preclinical study results are also used to determine the no-observable-adverse-effect level
(NOAEL) and estimate the “Maximum Safe Clinical Starting Dose” which are then used for
determining the dose range to be tested in the Phase I clinical trial1. Thus, the FDA depends
heavily on the preclinical pharmacology-toxicology program to support safety and plan the
Phase I clinical trial. Results of the preclinical pharmacology-toxicology program are submitted
to the FDA as part of an Investigational New Drug (IND) Application to support the proposed

                                                                                                    1
first-in-human clinical trial. The focus of the FDA’s clinical reviewers when assessing a Phase I
IND application is whether the first-in-human clinical study is designed to demonstrate safety in
a small number of subjects without putting these subjects at unnecessary risk.

Although safety is the main focus in the preclinical studies and early clinical trials, the sponsor
should also be thinking in terms of defining the relationship between dose, exposure and
efficacy. Historically, one of the main reasons for the high rate of drug attrition in clinical
development was undesirable pharmacokinetics (PK); from 1964 to 1985, approximately 39% of
drug candidates in the UK failed clinical development due to their PK characteristics2. This is no
longer a major issue due to improved approaches to drug design and candidate selection
increasing the bioavailability of new chemical entities. More recently, Phase 2 clinical trials
most often fail because insufficient attention was paid to accurately translating preclinical
efficacy findings to clinical doses that are not just safe, but have a high chance of demonstrating
efficacy3,4. In fact, the incidence of failure due to efficacy in Phase 2 clinical trials is actually
increasing based on an analysis of 2008-2010 data. Figure 1, drawn from a recent article by
Arrowsmith (2011)3, indicates that 51% of 87 Phase 2 trials conducted between 2008 and 2010
clearly failed because of efficacy. The authors report that an additional ~ 29% were
categorized as failures due to “strategic” reasons that likely resulted from failures due to
efficacy (as well as safety). Thus, the overwhelming majority of Phase 2 failures were related to
efficacy.

          60                                                     Figure 1. Phase 2 Trial Failures: 2008-
                 51                                              2010. Revised from Arrowsmith et
          50                                                     al.(2011)3 reporting data from 108 failed
                                                                 Phase 2 clinical trials. The reasons for
                                                                 failure were reported for 87 of the 108
          40
                                                                 failed trials and presented as the
                                                                 percentage of the failures in each
                             29
          30                                                     category. The numbers above each bar
Percent




                                                                 are the percentages.
                                             19
          20


          10

                                                        1
           0
               Efficacy   Strategic        Safety   PK/Biovail
                             Reasons for Failure




Analysis of Phase 3 trial failures between 2003-2007 indicate that ~45% are unsuccessful
because of failure to demonstrate efficacy compared to placebo5. These data for Phase 2 & 3
failures are very sobering to CEOs and investors when considering the prospects for success

                                                                                                         2
and return on investment. Looking at the top 50 companies and all classes of drugs, it is
estimated that approximately 1 drug in every 6 candidates that enter clinical testing receives
approval by FDA6. Since most of these drugs fail at the level of Phase 2 clinical testing, it is
critical that companies employ strategies that increase the chances of obtaining efficacy. This
will provide a much stronger basis for undertaking the major commitment of time and resources
required for Phase 3 clinical testing and reduce failures due to efficacy. An extremely important
part of this strategy is dose optimization, which represents one of the main causes of efficacy-
related failures in late-phase clinical testing. In fact, the FDA guidance on end-of-Phase 2A
meetings7 specifically states: “Accurate dose-response information is important for
understanding how patients should take drugs to maximize desirable effects and minimize
undesirable effects. Dose selection for Phase 2 and Phase 3 trials is a challenge in many drug
development programs, and poor choice may lead to trial failure. Improving early dose selection
may increase the likelihood of future trial success”.

It is estimated that no more than 8% of drugs entering clinical testing for oncology indications
ever receive licensure. This dismal record is viewed by some as simply unsustainable. To help
address these issues, PK data should be analyzed in tandem with pharmacodynamic (PD) data
to assess the pharmacokinetic/pharmacodynamic (PK/PD) relationship throughout the entire
drug development process. We address this problem early in preclinical and clinical
development, and are providing sponsors with the translational pharmacology resources to
successfully optimize their chances for success in their critical Phase 2 and Phase 3 clinical
trials.

Preclinical and Clinical Pharmacokinetic/Pharmacodynamic Studies

We view preclinical and clinical PK/PD studies as a continuum that permits optimal translation of
dose from animal studies to clinical trials and finally to clinical practice. PK/PD analysis should
not be performed as an afterthought or simply to meet regulatory requirements, but rather must
be carried out with careful planning from early development through product approval. The
fundamental principle of translational pharmacology is to design pharmacokinetic and
toxicokinetic studies in the preclinical setting and early Phase 1 clinical trials with the purpose of
accurately and effectively modeling the dosing so that critical clinical trials maximize their
chance of success with respect to both safety and efficacy. Therefore, the goal of translational
pharmacology is not simply to design preclinical studies to demonstrate safety for first-in-human
clinical administration, but to design studies that, together with Phase 1 clinical data, will be
used to maximize the chances of success in the Phase 2 and Phase 3 clinical trials. It is worth
noting that preclinical, first-in-human and other Phase 1 studies can be particularly well suited to
PK/PD analyses since a wide range of dose levels are often assessed and blood sampling
tends to be intensive (data rich). Furthermore, depending on the therapeutic area, biomarker
data can be incorporated into such studies relatively easily and biomarkers can play a role in
bridging animal and human pharmacology, toxicity/safety evaluation, dose selection, patient
selection. The use of biomarkers can be an integral part of reducing the risk of Phase 2 trial

                                                                                                    3
failure8. Later in clinical development, we utilize data gathered across clinical trials to
characterize the relationships between dose, safety, efficacy, biomarkers and key population
covariates. These data are used to help define dosing guidelines for use in clinical practice
following approval. Biomarkers have played a key role in accelerated approval.9

The following case study illustrates the need and critical role that PK/PD assessments and
modeling can play in increasing the chances for success in the development process. This
particular example is for an oncology drug, a therapeutic area which has one of the highest
failure rates (estimated at ≥ 90%10).

Case Study

A new biopharmaceutical for treating cancer has shown evidence of efficacy in both in vitro cell
culture studies and in vivo xenograft models of pancreatic tumors, including evidence for
synergism with both first line and second line therapeutic agents. Toxicology and toxicokinetic
data suggested that this new biopharmaceutical may have a very favorable adverse event
profile since it produced severe toxicity only at very high doses of drug. FDA allowed this drug to
enter a first-in-human Phase 1 clinical trial in advanced pancreatic cancer patients and the
results showed it to be well tolerated suggesting that this new agent was safe and could be
tested in a Phase 2 dose escalation trial. This Phase 1 trial demonstrated significant evidence
of safety at the doses tested and the investigators felt it was not necessary to add additional
dosing cohorts in an attempt to establish the maximum tolerable dose.

A Phase 2 clinical trial followed from this study and targeted pancreatic cancer patients based
on efficacy that was demonstrated in in vitro and in vivo proof-of-concept pharmacology studies
and the good tolerability and safety profile from the Phase 1 trial. The Phase 2 trial confirmed
the tolerability and safety profile of the drug, but was stopped because of futility to demonstrate
efficacy based on the prospective frequentist (non-Bayesian) design of the study (Figure 2.).

Figure 2 A Common Scenario

 Strong In vitro and         Excellent safety         Well tolerated in             Phase 2 trial
 in vivo evidence of       profile in toxicology      Phase 1 clinical           stopped for futility
       efficacy                   studies                    trial




Retrospective analysis of the PK data from the mouse xenograft models and the human PK
data from both the Phase 1 and 2 trials revealed that exposures (AUCs) were considerably
lower in humans than exposures observed with the pancreatic tumor lines that had
demonstrated efficacy in xenograft models. In fact, the results of this analysis indicated that the
levels of exposure attained in humans correlated with exposures seen in mouse models
showing slightly less efficacy in colon cancer cell lines. These data suggested that the wrong
type of tumor (pancreatic vs. colon) may have been selected for clinical investigation. Originally,

                                                                                                        4
the basis for selection of the disease (particular tumor) for study in clinical trials was the tumor
that showed the highest percent tumor growth suppression (Emax) in xenograft mouse models in
the absence of significant toxicity. However, this selection was premature and ignored the
difficulty of translating exposure in animal models to that achievable in humans. The selection
should have been based on a comparison (using appropriate allometric corrections) of the
exposures achieved in the Phase 1 clinical trial and those observed in preclinical studies
demonstrating efficacy in xenograft tumor models.

Additionally, this retrospective analysis of the data revealed that increasing the maximum dose
used in the Phase 1 clinical trial could have achieved the exposures associated with efficacy in
the xenograft mouse models of pancreatic cancer. Because the appropriate analyses had not
been performed on the preclinical and Phase 1 PK/PD data, the design of the Phase 2 trial did
not target a potentially efficacious dose. The investigators did not realize that further dose
escalation in the Phase 1 trial may have resulted in the selection of a dose for the Phase 2
study with a higher chance of demonstrating disease activity.

All too often the focus is solely on establishing the MTD in Phase 1 clinical trials and the
assumption that maybe some indications of potential efficacy will be obtained. While the number
of patients in a Phase 1 trial is not sufficient to demonstrate statistical evidence of efficacy, it is
important to establish that the exposures achieved in patients are in the efficacious range
established in the preclinical models. In addition, assessments of biomarkers can provide further
evidence of the potential for efficacy and make the transition to Phase 2 clinical trials more
successful.

Fortunately, in this particular case, the preclinical pharmacokinetic studies were designed such
that complete pharmacokinetic data were obtained allowing for robust estimates of the
relationship between efficacy and exposure to the test drug. These preclinical data, combined
with the human PK data, could thus be used to select the optimal tumor for further clinical study.
In addition, PK/PD modeling was used to determine the optimal study design for the next clinical
study, in particular the dosing range required to maximize the chances of observing efficacy
(Figure 3.). In this case, a Bayesian design with appropriate interim analyses was selected to
most efficiently show efficacy in the second Phase 2 study.




                                                                                                     5
Figure 3. Utilizing PK/PD to Achieve Accurate Translation of Efficacious Dosing

        Preclinical              Pharmacokinetic
        Efficacy in               Data (Cmax,
                                     AUC)                      Select tumor
     Xenograft models                                           for clinical         Phase 2 trial
                                                               study based             targeting
                                                               on exposure            exposures
       Phase 1&2                 Pharmacokinetic
       Clinical Trial             Data (Cmax,
                                     AUC)


                                   Pharmacokinetic/Pharmacodynamic Modeling



The data in Figure 4a show the plasma concentrations predicted for two concentrations of drug
to be used in the Phase 2 clinical trial including the 95% prediction intervals for these individual
plasma concentrations. Data like these can be not only important to establishing exposures that

Figure 4a.




                                                                                                     6
are likely to be efficacious, they also help to warn against the possibility of achieving drug
exposures in the toxic range for these patient populations. When the data was combined with
the reduction in expression of the tumor biomarker from the mouse xenograft models, it was
possible to predict the reduction in the appearance of this biomarker in patient plasma as shown
in Figure 4b. The data in Figure 4b show the predicted pharmacodynamics response for single
dose treatment at two different doses (200 and 600 mg) of drug.

Figure 4b.




With the actual PK and PD data from patients it is possible, using the Bayesian approach, to
select the optimal dose as well as the most susceptible/responsive patient subpopulations for
expansion in the Phase 2 clinical trial. The Bayesian approach utilizes prior data and
assessments, in this case the Phase 1 study patient PK data and data from the ongoing
Phase 2 trial patients, to provide better estimates of the treatment efficacy using biomarker
assessments. This approach continually learns and improves as the trial progresses to target
the optimal dose and most susceptible patient population in the Phase 2 clinical trials and
ultimately for testing in one or more Phase 3 clinical trials.




                                                                                                7
Commentary on the Case Study

This example illustrates how: 1) insufficient attention to PK/PD analysis and modeling all too
often contributes to the failure of Phase 2 trials and; 2) careful planning and study design that
results in robust PK/PD data from preclinical and Phase 1 clinical studies is critical to designing
successful Phase 2 and 3 clinical trials. For small companies, efforts to move forward with
additional clinical studies following failure of a Phase 2 clinical trial can be associated with
significant difficulties and delays that often prove unsustainable. At a minimum, one can expect
difficulty convincing investors to fund further studies due to the loss of confidence in the
candidate drug after a Phase 2 trial has been stopped due to futility. This loss of confidence is
also seen in the reduced enthusiasm of key opinion leaders and clinical investigators to
participate in further clinical testing. Furthermore, regulatory agencies may become much more
critical about what will be required for a sponsor to proceed with further clinical trials when
efficacy has been brought into question in the clinical setting. The assumption of efficacy based
on the preclinical data becomes a weaker argument for the efficacy potential of a product once it
is in clinical trials. These are extraordinarily difficult barriers to overcome and, at a minimum,
will constitute significant program delays and unplanned additional costs. For small companies,
practically speaking, there is really only one chance to get this right.

For small biotech companies, in particular, it is critical to establish integrated program design
from the beginning that optimizes the use of all prior data to avoid the myriad of pitfalls and
target successful clinical trials. This approach also permits the design of a development
program with the fewest number of studies and at the same time highest probability to succeed,
saving time, resources, and optimizing investor dollars.

Discussion

The importance of preclinical and clinical pharmacokinetics to designing a successful clinical
trial design cannot be overemphasized. The expense associated with PK/PD analysis and
modeling is trivial compared to the cost of failed clinical trials and the potentially devastating
consequences for small companies. Therefore, the careful planning of a translational
pharmacology program that spans preclinical through clinical studies and provides information
that maximizes the chances of success in the clinic is a service that adds great value to a
clinical development program for a biopharmaceutical or drug.

Accordingly, our approach is to establish a program that utilizes state-of-the-art PK/PD analysis
and modeling that will target and optimize the likelihood of demonstrating efficacy as early as
possible in clinical testing. Application of pharmacometrics across the entire development life
cycle is critical to: 1) the design and execution of a preclinical pharmacology-toxicology
program; 2) the design and execution of successful clinical trials; 3) achieving a positive
benefit/risk balance supporting licensure; 4) and establishing an effective post-marketing and

                                                                                                     8
pharmacovigilance program. Thus, PK/PD models are becoming increasingly critical
knowledge-building tools, not only for late phase clinical trials, but throughout the entire drug
development process.

Biologics Consulting Group and Quanticate welcome the opportunity to bring their combined
unparalleled depth and breadth of experience to navigate the major challenges inherent in the
development pathway to new therapeutic drug/biopharmaceutical licensure.




With over 18 years of experience with FDA registration of Biologics, Drugs, and Devices, BCG
has contributed to 10 of the last 12 BLA approvals by FDA. BCG supports product development
from pre-clinical pharmacology through clinical design and product registration. To find out how
we can support your programs, please visit us at www.biologicsconsulting.com.

Quanticate’s strong Biostatistical heritage and depth of experience offers statistical consultancy
in novel areas such as adaptive design methodology, PKPD modeling, trial design, DSMB/DMC,
and phase I-IV clinical and safety data analysis. To find out how Quanticate can augment your
internal trial teams please visit us at www.quanticate.com.

To learn more about how we can help you please contact the following individuals:

L. Bruce Pearce, Ph.D.                        Jimmy Brown
Senior Consultant                             Business Development Manager
Biologics Consulting Group, Inc.              Quanticate Ltd
office: 603-577-1646                          office: 857 998 6860
mobile: 603-809-9095                          mobile:617 800 4812
bpearce@bcg-usa.com                           james.brown@quanticate.com


Bibliography
1
  FDA Guidance for Industry. Estimating the maximum safe starting dose in Initial clinical trials for
   therapeutics in adult healthy volunteers. July 2005
2
  Prentis, R. A., Lisand, Y., Walker, S.R. Pharmaceutical innovation by the seven UK-owned
   pharmaceutical companies (1964-1985). J. Clin. Pharmacol. 25, 387-396,1988
3
  Arrowsmith, J, Phase II failures: 2008-2010. Nature Review [Drug Discovery] 10:May 2011
4
  Rubio-Viqueria B and Hidalgo M, Mouse xenograft models for drug discovery in pancreatic cancer.
   Chapter 2 In Drug Discovery and Pancreatic Cancer H. Han, P. Grippo (eds.) Springer Science +
   Business Media LLC,p 29-49, 2010.
5
  Invention Reinvented: McKinsey Perspectives on Pharmaceutical R&D, McKiney and Company, 2010
6
  Pharmaceutical Research and Manufacturers of America, PhRMA Pharmaceutical Industry Profile 2011
   (Washington, DC: 2011).
7
  FDA Guidance for Industry: End-of-Phase 2A Meetings, 2009
8
  Hurko, O, The uses of biomarkers in drug development. Ann. N.Y. Acad. Sci. 1180: 1–10, 2009

                                                                                                    9
9
    FDA Guidance for Industry. Clinical trial endpoints for the approval of cancer drugs and biologics, 2007
10
     Dimond, P F What’s wrong with phase II trials? Genetic Engineering and Biotechnology News. Feb 28,
    2011.




                                                                                                          10

Más contenido relacionado

La actualidad más candente

CPCSEA guidelines for lab animal facility
CPCSEA guidelines for lab animal facility CPCSEA guidelines for lab animal facility
CPCSEA guidelines for lab animal facility deepalisanap31
 
Screening of hepatoprotective drugs
Screening of hepatoprotective drugsScreening of hepatoprotective drugs
Screening of hepatoprotective drugsDipanjaliKamthe
 
STATISTICAL METHODS FOR PHARMACOVIGILANCE
STATISTICAL METHODS FOR PHARMACOVIGILANCESTATISTICAL METHODS FOR PHARMACOVIGILANCE
STATISTICAL METHODS FOR PHARMACOVIGILANCESanju Kaladharan
 
Screening Methods of Antihypertensive Agents
Screening Methods of Antihypertensive AgentsScreening Methods of Antihypertensive Agents
Screening Methods of Antihypertensive AgentsDr. Advaitha MV
 
ROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTION
ROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTIONROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTION
ROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTIONAnindya Banerjee
 
Importance and application of toxicokinetics
Importance and application of toxicokineticsImportance and application of toxicokinetics
Importance and application of toxicokineticsPrajjwal Rajput
 
Methods and Tools for ADR Reporting.pptx
Methods and Tools for ADR Reporting.pptxMethods and Tools for ADR Reporting.pptx
Methods and Tools for ADR Reporting.pptxPankajKadyan5
 
Translational pharmacology
Translational pharmacologyTranslational pharmacology
Translational pharmacologyRewariBhavya
 
DESIGN OF DOSAGE REGIMEN.pptx
DESIGN OF DOSAGE REGIMEN.pptxDESIGN OF DOSAGE REGIMEN.pptx
DESIGN OF DOSAGE REGIMEN.pptxDrAniqaSundas
 
Aris G PHARMACOVIGILANCE AND VIGIFLOW
Aris G PHARMACOVIGILANCE AND VIGIFLOWAris G PHARMACOVIGILANCE AND VIGIFLOW
Aris G PHARMACOVIGILANCE AND VIGIFLOWayanarkumar19
 
TDM of drugs used in organ transplantation
TDM of drugs used in organ transplantationTDM of drugs used in organ transplantation
TDM of drugs used in organ transplantationDr. Ramesh Bhandari
 
Documentation clinical trial
Documentation clinical trialDocumentation clinical trial
Documentation clinical trialankit sharma
 
Genetic polymorphism in drug transport and drug targets.
Genetic polymorphism in drug transport and drug targets.Genetic polymorphism in drug transport and drug targets.
Genetic polymorphism in drug transport and drug targets.pavithra vinayak
 
BUILDING BLOCKS & evaluation process in qum.pptx
BUILDING BLOCKS  & evaluation process in qum.pptxBUILDING BLOCKS  & evaluation process in qum.pptx
BUILDING BLOCKS & evaluation process in qum.pptxAmeena Kadar
 
Regulatory terminologies used in PV (Pharmacovigilance)
Regulatory terminologies used in PV (Pharmacovigilance)Regulatory terminologies used in PV (Pharmacovigilance)
Regulatory terminologies used in PV (Pharmacovigilance)MubasheeraMg
 
Suhas (chronopharmacology of diabetes)
Suhas (chronopharmacology of diabetes)Suhas (chronopharmacology of diabetes)
Suhas (chronopharmacology of diabetes)suhaspatil114
 
Adr reporting ppt
Adr reporting pptAdr reporting ppt
Adr reporting pptRimaSingh14
 
Therapeutic drug monitoring of cardiovascular drugs
Therapeutic drug monitoring of cardiovascular drugsTherapeutic drug monitoring of cardiovascular drugs
Therapeutic drug monitoring of cardiovascular drugsDr. Ramesh Bhandari
 

La actualidad más candente (20)

CPCSEA guidelines for lab animal facility
CPCSEA guidelines for lab animal facility CPCSEA guidelines for lab animal facility
CPCSEA guidelines for lab animal facility
 
Screening of hepatoprotective drugs
Screening of hepatoprotective drugsScreening of hepatoprotective drugs
Screening of hepatoprotective drugs
 
STATISTICAL METHODS FOR PHARMACOVIGILANCE
STATISTICAL METHODS FOR PHARMACOVIGILANCESTATISTICAL METHODS FOR PHARMACOVIGILANCE
STATISTICAL METHODS FOR PHARMACOVIGILANCE
 
Screening Methods of Antihypertensive Agents
Screening Methods of Antihypertensive AgentsScreening Methods of Antihypertensive Agents
Screening Methods of Antihypertensive Agents
 
ROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTION
ROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTIONROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTION
ROLE OF PHARMACOVIGILANCE AGAINST ADVERSE DRUG REACTION
 
Importance and application of toxicokinetics
Importance and application of toxicokineticsImportance and application of toxicokinetics
Importance and application of toxicokinetics
 
Methods and Tools for ADR Reporting.pptx
Methods and Tools for ADR Reporting.pptxMethods and Tools for ADR Reporting.pptx
Methods and Tools for ADR Reporting.pptx
 
Translational pharmacology
Translational pharmacologyTranslational pharmacology
Translational pharmacology
 
DESIGN OF DOSAGE REGIMEN.pptx
DESIGN OF DOSAGE REGIMEN.pptxDESIGN OF DOSAGE REGIMEN.pptx
DESIGN OF DOSAGE REGIMEN.pptx
 
Aris G PHARMACOVIGILANCE AND VIGIFLOW
Aris G PHARMACOVIGILANCE AND VIGIFLOWAris G PHARMACOVIGILANCE AND VIGIFLOW
Aris G PHARMACOVIGILANCE AND VIGIFLOW
 
TDM of drugs used in organ transplantation
TDM of drugs used in organ transplantationTDM of drugs used in organ transplantation
TDM of drugs used in organ transplantation
 
Documentation clinical trial
Documentation clinical trialDocumentation clinical trial
Documentation clinical trial
 
Genetic polymorphism in drug transport and drug targets.
Genetic polymorphism in drug transport and drug targets.Genetic polymorphism in drug transport and drug targets.
Genetic polymorphism in drug transport and drug targets.
 
BUILDING BLOCKS & evaluation process in qum.pptx
BUILDING BLOCKS  & evaluation process in qum.pptxBUILDING BLOCKS  & evaluation process in qum.pptx
BUILDING BLOCKS & evaluation process in qum.pptx
 
Regulatory terminologies used in PV (Pharmacovigilance)
Regulatory terminologies used in PV (Pharmacovigilance)Regulatory terminologies used in PV (Pharmacovigilance)
Regulatory terminologies used in PV (Pharmacovigilance)
 
Suhas (chronopharmacology of diabetes)
Suhas (chronopharmacology of diabetes)Suhas (chronopharmacology of diabetes)
Suhas (chronopharmacology of diabetes)
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
 
Adr reporting ppt
Adr reporting pptAdr reporting ppt
Adr reporting ppt
 
Therapeutic drug monitoring of cardiovascular drugs
Therapeutic drug monitoring of cardiovascular drugsTherapeutic drug monitoring of cardiovascular drugs
Therapeutic drug monitoring of cardiovascular drugs
 
CRO.pptx
CRO.pptxCRO.pptx
CRO.pptx
 

Similar a Translational pharmacology

Challenges in Phase III Cancer Clinical Trials
Challenges in Phase III Cancer Clinical Trials Challenges in Phase III Cancer Clinical Trials
Challenges in Phase III Cancer Clinical Trials Bhaswat Chakraborty
 
Drug Discovery Today v9p976
Drug Discovery Today v9p976Drug Discovery Today v9p976
Drug Discovery Today v9p976Michael Weiner
 
Biomarker roles within clinical trials
Biomarker roles within clinical trialsBiomarker roles within clinical trials
Biomarker roles within clinical trialsRay Wright
 
Phases of clinical trial 11.9.14
Phases of clinical trial 11.9.14Phases of clinical trial 11.9.14
Phases of clinical trial 11.9.14DR ANUP PETARE
 
Toxicology screening and therapeutic drug monitoring (an introduction)
 Toxicology screening and therapeutic drug monitoring (an introduction)  Toxicology screening and therapeutic drug monitoring (an introduction)
Toxicology screening and therapeutic drug monitoring (an introduction) Hossamaldin Alzawawi
 
Stages of drug development
Stages of drug developmentStages of drug development
Stages of drug developmentmeethy
 
Potential of phase II clinical trials in drug development
Potential of phase II clinical trials in drug developmentPotential of phase II clinical trials in drug development
Potential of phase II clinical trials in drug developmentBhaswat Chakraborty
 
Pharmacology/Toxicology information to submit an IND for an anticancer drug
Pharmacology/Toxicology information to submit an IND for an anticancer drugPharmacology/Toxicology information to submit an IND for an anticancer drug
Pharmacology/Toxicology information to submit an IND for an anticancer drugshabeel pn
 
Clinical trial phases, requirements and regulations
Clinical trial  phases, requirements and regulationsClinical trial  phases, requirements and regulations
Clinical trial phases, requirements and regulationsDr. Siddhartha Dutta
 
Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...
Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...
Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...Rosmirella Cano Rojas
 
CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...
CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...
CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...Clarinda Cerejo
 
Pharmaceutical product development and its associated quality system 01
Pharmaceutical product development and its associated quality system 01Pharmaceutical product development and its associated quality system 01
Pharmaceutical product development and its associated quality system 01Abdirizak Mohammed
 
Pharmacometrics 2.2.17
Pharmacometrics 2.2.17Pharmacometrics 2.2.17
Pharmacometrics 2.2.17Dr. Md Yaqub
 
Pharmacometrics 2.2.17
Pharmacometrics 2.2.17Pharmacometrics 2.2.17
Pharmacometrics 2.2.17Dr. Md Yaqub
 
Decoding Phase II Clinical Trial Failures
Decoding Phase II Clinical Trial FailuresDecoding Phase II Clinical Trial Failures
Decoding Phase II Clinical Trial Failuressubhabbasu
 

Similar a Translational pharmacology (20)

Challenges in Phase III Cancer Clinical Trials
Challenges in Phase III Cancer Clinical Trials Challenges in Phase III Cancer Clinical Trials
Challenges in Phase III Cancer Clinical Trials
 
Drug Discovery Today v9p976
Drug Discovery Today v9p976Drug Discovery Today v9p976
Drug Discovery Today v9p976
 
Biomarker roles within clinical trials
Biomarker roles within clinical trialsBiomarker roles within clinical trials
Biomarker roles within clinical trials
 
PTSM 2.pptx
PTSM 2.pptxPTSM 2.pptx
PTSM 2.pptx
 
Phases of clinical trial 11.9.14
Phases of clinical trial 11.9.14Phases of clinical trial 11.9.14
Phases of clinical trial 11.9.14
 
presentation.pptx
presentation.pptxpresentation.pptx
presentation.pptx
 
Toxicology screening and therapeutic drug monitoring (an introduction)
 Toxicology screening and therapeutic drug monitoring (an introduction)  Toxicology screening and therapeutic drug monitoring (an introduction)
Toxicology screening and therapeutic drug monitoring (an introduction)
 
EOP.SOJA.S5
EOP.SOJA.S5EOP.SOJA.S5
EOP.SOJA.S5
 
Stages of drug development
Stages of drug developmentStages of drug development
Stages of drug development
 
Potential of phase II clinical trials in drug development
Potential of phase II clinical trials in drug developmentPotential of phase II clinical trials in drug development
Potential of phase II clinical trials in drug development
 
Pharmacology/Toxicology information to submit an IND for an anticancer drug
Pharmacology/Toxicology information to submit an IND for an anticancer drugPharmacology/Toxicology information to submit an IND for an anticancer drug
Pharmacology/Toxicology information to submit an IND for an anticancer drug
 
CLINICAL TRIALS.pptx
CLINICAL TRIALS.pptxCLINICAL TRIALS.pptx
CLINICAL TRIALS.pptx
 
Clinical trial phases, requirements and regulations
Clinical trial  phases, requirements and regulationsClinical trial  phases, requirements and regulations
Clinical trial phases, requirements and regulations
 
Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...
Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...
Biosimilaridad e intercambiabilidad: principios y evidencia: una revisión sis...
 
CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...
CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...
CONSISTENCY OF CLINICAL DATA REPORTING BETWEEN CLINICALTRIALS.GOV AND PUBLICA...
 
Pharmaceutical product development and its associated quality system 01
Pharmaceutical product development and its associated quality system 01Pharmaceutical product development and its associated quality system 01
Pharmaceutical product development and its associated quality system 01
 
Pharmacometrics 2.2.17
Pharmacometrics 2.2.17Pharmacometrics 2.2.17
Pharmacometrics 2.2.17
 
Pharmacometrics 2.2.17
Pharmacometrics 2.2.17Pharmacometrics 2.2.17
Pharmacometrics 2.2.17
 
Phase I trials - Educational course at the PAMM-2019 winter meeting
Phase I trials - Educational course at the PAMM-2019 winter meetingPhase I trials - Educational course at the PAMM-2019 winter meeting
Phase I trials - Educational course at the PAMM-2019 winter meeting
 
Decoding Phase II Clinical Trial Failures
Decoding Phase II Clinical Trial FailuresDecoding Phase II Clinical Trial Failures
Decoding Phase II Clinical Trial Failures
 

Translational pharmacology

  • 1. Make life simpler Translational Pharmacology: The Key to Ensuring Safe and Effective Dosing L. Bruce Pearce, Ph.D1., Sue McKendrick, MSc, CStat2, John J. Jessop, Ph.D. 1, Julia Barrett, M.D. 1 and Mark Baird, BSc(Hons)2 1 Biologics Consulting Group, Inc. Alexandria VA, 2Quanticate, Ltd. Hitchin, UK Introduction: The relationship between dose, systemic exposure, and both safety and efficacy are the most important elements in biopharmaceutical/drug development. The key is to attain adequate systemic exposure to mediate a clinically effective response while avoiding a level of exposure that results in toxicity. Understanding these relationships constitutes the central dogma of product development from preclinical studies and on through to Phase 1, 2, and 3 clinical trials, and is still important in post-marketing studies and pharmacovigilance. In order to proceed successfully through clinical development, it is necessary to accurately assess and demonstrate a favorable risk/benefit relationship at each milestone. This begins at the preclinical level, where appropriate pharmacodynamic, safety pharmacology, pharmacokinetic, toxicology and toxicokinetic studies are performed in various animal species. The results of these studies are used to demonstrate proof-of-concept for the proposed indication and to support safety for first-in-human administration in a Phase I clinical trial. These preclinical study results are also used to determine the no-observable-adverse-effect level (NOAEL) and estimate the “Maximum Safe Clinical Starting Dose” which are then used for determining the dose range to be tested in the Phase I clinical trial1. Thus, the FDA depends heavily on the preclinical pharmacology-toxicology program to support safety and plan the Phase I clinical trial. Results of the preclinical pharmacology-toxicology program are submitted to the FDA as part of an Investigational New Drug (IND) Application to support the proposed 1
  • 2. first-in-human clinical trial. The focus of the FDA’s clinical reviewers when assessing a Phase I IND application is whether the first-in-human clinical study is designed to demonstrate safety in a small number of subjects without putting these subjects at unnecessary risk. Although safety is the main focus in the preclinical studies and early clinical trials, the sponsor should also be thinking in terms of defining the relationship between dose, exposure and efficacy. Historically, one of the main reasons for the high rate of drug attrition in clinical development was undesirable pharmacokinetics (PK); from 1964 to 1985, approximately 39% of drug candidates in the UK failed clinical development due to their PK characteristics2. This is no longer a major issue due to improved approaches to drug design and candidate selection increasing the bioavailability of new chemical entities. More recently, Phase 2 clinical trials most often fail because insufficient attention was paid to accurately translating preclinical efficacy findings to clinical doses that are not just safe, but have a high chance of demonstrating efficacy3,4. In fact, the incidence of failure due to efficacy in Phase 2 clinical trials is actually increasing based on an analysis of 2008-2010 data. Figure 1, drawn from a recent article by Arrowsmith (2011)3, indicates that 51% of 87 Phase 2 trials conducted between 2008 and 2010 clearly failed because of efficacy. The authors report that an additional ~ 29% were categorized as failures due to “strategic” reasons that likely resulted from failures due to efficacy (as well as safety). Thus, the overwhelming majority of Phase 2 failures were related to efficacy. 60 Figure 1. Phase 2 Trial Failures: 2008- 51 2010. Revised from Arrowsmith et 50 al.(2011)3 reporting data from 108 failed Phase 2 clinical trials. The reasons for failure were reported for 87 of the 108 40 failed trials and presented as the percentage of the failures in each 29 30 category. The numbers above each bar Percent are the percentages. 19 20 10 1 0 Efficacy Strategic Safety PK/Biovail Reasons for Failure Analysis of Phase 3 trial failures between 2003-2007 indicate that ~45% are unsuccessful because of failure to demonstrate efficacy compared to placebo5. These data for Phase 2 & 3 failures are very sobering to CEOs and investors when considering the prospects for success 2
  • 3. and return on investment. Looking at the top 50 companies and all classes of drugs, it is estimated that approximately 1 drug in every 6 candidates that enter clinical testing receives approval by FDA6. Since most of these drugs fail at the level of Phase 2 clinical testing, it is critical that companies employ strategies that increase the chances of obtaining efficacy. This will provide a much stronger basis for undertaking the major commitment of time and resources required for Phase 3 clinical testing and reduce failures due to efficacy. An extremely important part of this strategy is dose optimization, which represents one of the main causes of efficacy- related failures in late-phase clinical testing. In fact, the FDA guidance on end-of-Phase 2A meetings7 specifically states: “Accurate dose-response information is important for understanding how patients should take drugs to maximize desirable effects and minimize undesirable effects. Dose selection for Phase 2 and Phase 3 trials is a challenge in many drug development programs, and poor choice may lead to trial failure. Improving early dose selection may increase the likelihood of future trial success”. It is estimated that no more than 8% of drugs entering clinical testing for oncology indications ever receive licensure. This dismal record is viewed by some as simply unsustainable. To help address these issues, PK data should be analyzed in tandem with pharmacodynamic (PD) data to assess the pharmacokinetic/pharmacodynamic (PK/PD) relationship throughout the entire drug development process. We address this problem early in preclinical and clinical development, and are providing sponsors with the translational pharmacology resources to successfully optimize their chances for success in their critical Phase 2 and Phase 3 clinical trials. Preclinical and Clinical Pharmacokinetic/Pharmacodynamic Studies We view preclinical and clinical PK/PD studies as a continuum that permits optimal translation of dose from animal studies to clinical trials and finally to clinical practice. PK/PD analysis should not be performed as an afterthought or simply to meet regulatory requirements, but rather must be carried out with careful planning from early development through product approval. The fundamental principle of translational pharmacology is to design pharmacokinetic and toxicokinetic studies in the preclinical setting and early Phase 1 clinical trials with the purpose of accurately and effectively modeling the dosing so that critical clinical trials maximize their chance of success with respect to both safety and efficacy. Therefore, the goal of translational pharmacology is not simply to design preclinical studies to demonstrate safety for first-in-human clinical administration, but to design studies that, together with Phase 1 clinical data, will be used to maximize the chances of success in the Phase 2 and Phase 3 clinical trials. It is worth noting that preclinical, first-in-human and other Phase 1 studies can be particularly well suited to PK/PD analyses since a wide range of dose levels are often assessed and blood sampling tends to be intensive (data rich). Furthermore, depending on the therapeutic area, biomarker data can be incorporated into such studies relatively easily and biomarkers can play a role in bridging animal and human pharmacology, toxicity/safety evaluation, dose selection, patient selection. The use of biomarkers can be an integral part of reducing the risk of Phase 2 trial 3
  • 4. failure8. Later in clinical development, we utilize data gathered across clinical trials to characterize the relationships between dose, safety, efficacy, biomarkers and key population covariates. These data are used to help define dosing guidelines for use in clinical practice following approval. Biomarkers have played a key role in accelerated approval.9 The following case study illustrates the need and critical role that PK/PD assessments and modeling can play in increasing the chances for success in the development process. This particular example is for an oncology drug, a therapeutic area which has one of the highest failure rates (estimated at ≥ 90%10). Case Study A new biopharmaceutical for treating cancer has shown evidence of efficacy in both in vitro cell culture studies and in vivo xenograft models of pancreatic tumors, including evidence for synergism with both first line and second line therapeutic agents. Toxicology and toxicokinetic data suggested that this new biopharmaceutical may have a very favorable adverse event profile since it produced severe toxicity only at very high doses of drug. FDA allowed this drug to enter a first-in-human Phase 1 clinical trial in advanced pancreatic cancer patients and the results showed it to be well tolerated suggesting that this new agent was safe and could be tested in a Phase 2 dose escalation trial. This Phase 1 trial demonstrated significant evidence of safety at the doses tested and the investigators felt it was not necessary to add additional dosing cohorts in an attempt to establish the maximum tolerable dose. A Phase 2 clinical trial followed from this study and targeted pancreatic cancer patients based on efficacy that was demonstrated in in vitro and in vivo proof-of-concept pharmacology studies and the good tolerability and safety profile from the Phase 1 trial. The Phase 2 trial confirmed the tolerability and safety profile of the drug, but was stopped because of futility to demonstrate efficacy based on the prospective frequentist (non-Bayesian) design of the study (Figure 2.). Figure 2 A Common Scenario Strong In vitro and Excellent safety Well tolerated in Phase 2 trial in vivo evidence of profile in toxicology Phase 1 clinical stopped for futility efficacy studies trial Retrospective analysis of the PK data from the mouse xenograft models and the human PK data from both the Phase 1 and 2 trials revealed that exposures (AUCs) were considerably lower in humans than exposures observed with the pancreatic tumor lines that had demonstrated efficacy in xenograft models. In fact, the results of this analysis indicated that the levels of exposure attained in humans correlated with exposures seen in mouse models showing slightly less efficacy in colon cancer cell lines. These data suggested that the wrong type of tumor (pancreatic vs. colon) may have been selected for clinical investigation. Originally, 4
  • 5. the basis for selection of the disease (particular tumor) for study in clinical trials was the tumor that showed the highest percent tumor growth suppression (Emax) in xenograft mouse models in the absence of significant toxicity. However, this selection was premature and ignored the difficulty of translating exposure in animal models to that achievable in humans. The selection should have been based on a comparison (using appropriate allometric corrections) of the exposures achieved in the Phase 1 clinical trial and those observed in preclinical studies demonstrating efficacy in xenograft tumor models. Additionally, this retrospective analysis of the data revealed that increasing the maximum dose used in the Phase 1 clinical trial could have achieved the exposures associated with efficacy in the xenograft mouse models of pancreatic cancer. Because the appropriate analyses had not been performed on the preclinical and Phase 1 PK/PD data, the design of the Phase 2 trial did not target a potentially efficacious dose. The investigators did not realize that further dose escalation in the Phase 1 trial may have resulted in the selection of a dose for the Phase 2 study with a higher chance of demonstrating disease activity. All too often the focus is solely on establishing the MTD in Phase 1 clinical trials and the assumption that maybe some indications of potential efficacy will be obtained. While the number of patients in a Phase 1 trial is not sufficient to demonstrate statistical evidence of efficacy, it is important to establish that the exposures achieved in patients are in the efficacious range established in the preclinical models. In addition, assessments of biomarkers can provide further evidence of the potential for efficacy and make the transition to Phase 2 clinical trials more successful. Fortunately, in this particular case, the preclinical pharmacokinetic studies were designed such that complete pharmacokinetic data were obtained allowing for robust estimates of the relationship between efficacy and exposure to the test drug. These preclinical data, combined with the human PK data, could thus be used to select the optimal tumor for further clinical study. In addition, PK/PD modeling was used to determine the optimal study design for the next clinical study, in particular the dosing range required to maximize the chances of observing efficacy (Figure 3.). In this case, a Bayesian design with appropriate interim analyses was selected to most efficiently show efficacy in the second Phase 2 study. 5
  • 6. Figure 3. Utilizing PK/PD to Achieve Accurate Translation of Efficacious Dosing Preclinical Pharmacokinetic Efficacy in Data (Cmax, AUC) Select tumor Xenograft models for clinical Phase 2 trial study based targeting on exposure exposures Phase 1&2 Pharmacokinetic Clinical Trial Data (Cmax, AUC) Pharmacokinetic/Pharmacodynamic Modeling The data in Figure 4a show the plasma concentrations predicted for two concentrations of drug to be used in the Phase 2 clinical trial including the 95% prediction intervals for these individual plasma concentrations. Data like these can be not only important to establishing exposures that Figure 4a. 6
  • 7. are likely to be efficacious, they also help to warn against the possibility of achieving drug exposures in the toxic range for these patient populations. When the data was combined with the reduction in expression of the tumor biomarker from the mouse xenograft models, it was possible to predict the reduction in the appearance of this biomarker in patient plasma as shown in Figure 4b. The data in Figure 4b show the predicted pharmacodynamics response for single dose treatment at two different doses (200 and 600 mg) of drug. Figure 4b. With the actual PK and PD data from patients it is possible, using the Bayesian approach, to select the optimal dose as well as the most susceptible/responsive patient subpopulations for expansion in the Phase 2 clinical trial. The Bayesian approach utilizes prior data and assessments, in this case the Phase 1 study patient PK data and data from the ongoing Phase 2 trial patients, to provide better estimates of the treatment efficacy using biomarker assessments. This approach continually learns and improves as the trial progresses to target the optimal dose and most susceptible patient population in the Phase 2 clinical trials and ultimately for testing in one or more Phase 3 clinical trials. 7
  • 8. Commentary on the Case Study This example illustrates how: 1) insufficient attention to PK/PD analysis and modeling all too often contributes to the failure of Phase 2 trials and; 2) careful planning and study design that results in robust PK/PD data from preclinical and Phase 1 clinical studies is critical to designing successful Phase 2 and 3 clinical trials. For small companies, efforts to move forward with additional clinical studies following failure of a Phase 2 clinical trial can be associated with significant difficulties and delays that often prove unsustainable. At a minimum, one can expect difficulty convincing investors to fund further studies due to the loss of confidence in the candidate drug after a Phase 2 trial has been stopped due to futility. This loss of confidence is also seen in the reduced enthusiasm of key opinion leaders and clinical investigators to participate in further clinical testing. Furthermore, regulatory agencies may become much more critical about what will be required for a sponsor to proceed with further clinical trials when efficacy has been brought into question in the clinical setting. The assumption of efficacy based on the preclinical data becomes a weaker argument for the efficacy potential of a product once it is in clinical trials. These are extraordinarily difficult barriers to overcome and, at a minimum, will constitute significant program delays and unplanned additional costs. For small companies, practically speaking, there is really only one chance to get this right. For small biotech companies, in particular, it is critical to establish integrated program design from the beginning that optimizes the use of all prior data to avoid the myriad of pitfalls and target successful clinical trials. This approach also permits the design of a development program with the fewest number of studies and at the same time highest probability to succeed, saving time, resources, and optimizing investor dollars. Discussion The importance of preclinical and clinical pharmacokinetics to designing a successful clinical trial design cannot be overemphasized. The expense associated with PK/PD analysis and modeling is trivial compared to the cost of failed clinical trials and the potentially devastating consequences for small companies. Therefore, the careful planning of a translational pharmacology program that spans preclinical through clinical studies and provides information that maximizes the chances of success in the clinic is a service that adds great value to a clinical development program for a biopharmaceutical or drug. Accordingly, our approach is to establish a program that utilizes state-of-the-art PK/PD analysis and modeling that will target and optimize the likelihood of demonstrating efficacy as early as possible in clinical testing. Application of pharmacometrics across the entire development life cycle is critical to: 1) the design and execution of a preclinical pharmacology-toxicology program; 2) the design and execution of successful clinical trials; 3) achieving a positive benefit/risk balance supporting licensure; 4) and establishing an effective post-marketing and 8
  • 9. pharmacovigilance program. Thus, PK/PD models are becoming increasingly critical knowledge-building tools, not only for late phase clinical trials, but throughout the entire drug development process. Biologics Consulting Group and Quanticate welcome the opportunity to bring their combined unparalleled depth and breadth of experience to navigate the major challenges inherent in the development pathway to new therapeutic drug/biopharmaceutical licensure. With over 18 years of experience with FDA registration of Biologics, Drugs, and Devices, BCG has contributed to 10 of the last 12 BLA approvals by FDA. BCG supports product development from pre-clinical pharmacology through clinical design and product registration. To find out how we can support your programs, please visit us at www.biologicsconsulting.com. Quanticate’s strong Biostatistical heritage and depth of experience offers statistical consultancy in novel areas such as adaptive design methodology, PKPD modeling, trial design, DSMB/DMC, and phase I-IV clinical and safety data analysis. To find out how Quanticate can augment your internal trial teams please visit us at www.quanticate.com. To learn more about how we can help you please contact the following individuals: L. Bruce Pearce, Ph.D. Jimmy Brown Senior Consultant Business Development Manager Biologics Consulting Group, Inc. Quanticate Ltd office: 603-577-1646 office: 857 998 6860 mobile: 603-809-9095 mobile:617 800 4812 bpearce@bcg-usa.com james.brown@quanticate.com Bibliography 1 FDA Guidance for Industry. Estimating the maximum safe starting dose in Initial clinical trials for therapeutics in adult healthy volunteers. July 2005 2 Prentis, R. A., Lisand, Y., Walker, S.R. Pharmaceutical innovation by the seven UK-owned pharmaceutical companies (1964-1985). J. Clin. Pharmacol. 25, 387-396,1988 3 Arrowsmith, J, Phase II failures: 2008-2010. Nature Review [Drug Discovery] 10:May 2011 4 Rubio-Viqueria B and Hidalgo M, Mouse xenograft models for drug discovery in pancreatic cancer. Chapter 2 In Drug Discovery and Pancreatic Cancer H. Han, P. Grippo (eds.) Springer Science + Business Media LLC,p 29-49, 2010. 5 Invention Reinvented: McKinsey Perspectives on Pharmaceutical R&D, McKiney and Company, 2010 6 Pharmaceutical Research and Manufacturers of America, PhRMA Pharmaceutical Industry Profile 2011 (Washington, DC: 2011). 7 FDA Guidance for Industry: End-of-Phase 2A Meetings, 2009 8 Hurko, O, The uses of biomarkers in drug development. Ann. N.Y. Acad. Sci. 1180: 1–10, 2009 9
  • 10. 9 FDA Guidance for Industry. Clinical trial endpoints for the approval of cancer drugs and biologics, 2007 10 Dimond, P F What’s wrong with phase II trials? Genetic Engineering and Biotechnology News. Feb 28, 2011. 10