SlideShare a Scribd company logo
1 of 6
Download to read offline
Dr. Jan Trøst Jørgensen
THE “GATEKEEPER” OF PRECISION MEDICINE EVOLVES.
Drug-diagnostic codevelopment can lead to a streamlined approval strategy, but
will need to change in the future.
PHARMA & LIFE SCIENCES
INTERVIEW
Director, Dx-Rx Institute
2Elsevier R&D Solutions
“...packaging a diagnostic
with one drug may no longer
be realistic; instead, we may
need a networked approach
involving multiple drugs.”
Dr. Jørgensen has been involved in precision medicine since 1999 and has written
several thought-provoking articles on the topic. He has observed this paradigm shift
in pharmacotherapy roar through initial hype, wane in interest, and then settle into a
steady transformation in the development of new medicines built around the under-
standing that diseases and patient populations are heterogeneous. Dr. Jørgensen is
a consultant for drug developers and a driving force behind companion diagnostics,
which, in his own words, are “a kind of ‘gatekeeper’ in relation to which patients should
be treated with the drug in question.” In this interview, Dr. Jørgensen talks about the
implications of this gatekeeping role and the future of companion diagnostics.
As we learn more about the biology of
disease, the construct “one test, one
drug” will no longer be sustainable.
YOU ARE A STRONG PROPONENT OF THE DRUG-DIAGNOSTIC CODEVELOPMENT
MODEL WHERE COLLABORATION BETWEEN DEVELOPERS OF A DRUG AND
DEVELOPERS OF A DIAGNOSTIC THAT IDENTIFIES PATIENTS FOR THAT DRUG
RESULTS IN A COUPLED, AND AT TIMES EVEN SPEEDIER, APPROVAL OF BOTH.
WHAT DOES THIS MODEL LOOK LIKE IN PRACTICE?
This co-development model was inspired by the simultaneous FDA approval of
the breast cancer medication trastuzumab (Herceptin®, Roche/Genentech) and the
immunohistochemical assay for HER2 (HercepTest®, Dako) in 1998. It was also the
FDA that first initiated a discussion about drug-diagnostic codevelopment with a
2005 publication where they outlined several important issues for the establishment
of routine coupled approvals. Ideally, a drug developer begins working with an assay
developer during the discovery and preclinical phases of a drug development program
(Figure 1). Starting from a biomarker hypothesis that the assay developer uses to
establish a prototype diagnostic, there is a continuous exchange of information
between the two parties. The drug developer uses the prototype diagnostic to plan and
conduct early clinical trials. The assay developer uses information from clinical efforts
to assess the predictive characteristics of the biomarker and establish the appropriate
cut-off for patient inclusion, among other things. A continuous and structured dialog
between both parties is essential. It should be said, that this early collaboration and
iterative exchange is not always reality, however, it is the ideal relationship under which
a codevelopment becomes robust and has greater chances of success.
Discovery
Research
Pre-clinical
Drug Development
Diagnostic Development
Parallel Development of Drug and Diagnostic
Clinical
Phase I
Clinical
Phase II
Clinical
Phase III
Regulatory
Approval
Post
Approval
Phase
Biomarker
Selection
Feasibility
Studies
Prototype
Assay(s)
Analytical
Validation
Clinical
Validation
Regulatory
Approval
Clinical
Utility
Figure 1. The drug-diagnostic codevelopment model builds on a two-way dialog between drug and assay developers.
Early collaboration between the two parties coordinates development efforts for drug and diagnostic assay, which
results in a strong regulatory approval strategy.
Adapted from an original design by Dr. Jan Trøst Jørgensen.
3Elsevier RD Solutions
We have to bear in mind that companion diagnostics
is in its infancy and codevelopment programs
experience all the accompanying growing pains.
There is, for example, discord between the regulatory
status and consequent requirements of diagnostics
in general and the “gatekeeper” role of a companion
diagnostic. This is something Daniel Hayes from
the University of Michigan, Ann Arbor, and myself
have been pointing out for some time. Under our
current vision of stratified medicine, where a patient
population is subdivided — stratified — into groups
based on biological characteristics that are relevant
to an individual’s response to a drug, a companion
diagnostic is the gatekeeper that determines which
patients should be treated with a given drug. As this
pivotal node in a patient’s therapy, the diagnostic
assay greatly impacts therapeutic outcome and
should be subjected to the same level of regulatory
requirements as the drug it accompanies.
Having said that, the link between drug and
diagnostic at the point of therapy extends back to
the development and approval process, and this
can be very advantageous. Let’s start with the use
of the prototype diagnostic to plan and conduct
phase I and phase II clinical trials. Information from
these stages may feed back to further optimize a
compound. Also, the data from these trials are used
to establish the cut-off for patient inclusion in phase
III trials and, ultimately, clinical use after approval.
The right cut-off plays a critical role in the approval
strategy for the drug-diagnostic pair. This is a highly
complex analysis that can take months to complete
and balances relevant biology (hypothesized drug
mode of action, disease mechanisms, observed
response rates) with the sensitivity and specificity
of the diagnostic and several clinical trial design
aspects that impact demonstrating the efficacy of the
drug. In other words, the codevelopment leads to a
better informed and streamlined regulatory approval
strategy. Just 5 or 6 years ago, drug developers were
hesitant to engage early with diagnostic developers,
partly because of the uncertainty of success in drug
development. Sometime they would first contact a
diagnostic developer after not achieving the targeted
efficacy in phase II trials. Now we see companion
diagnostics entering the framework much earlier and
the result of this front-ended collaboration can be
a fast-tracked approval, like that of the lung cancer
drug crizotinib (Xalkori®, Pfizer) and its companion
diagnostic (VYSIS® ALK Break Apart FISH Probe
Kit, Abbott). Thus, uncertainty about a drug should
not stop you from working toward a companion
diagnostic, because it could rescue your drug!
THE REGULATORY APPROVAL OF A NEW DRUG IS A RESOURCE AND TIME-INTENSIVE ENDEAVOR, OFTEN
TAKING SEVERAL YEARS TO ACCOMPLISH. ADDING A DIAGNOSTIC ASSAY TO THE PICTURE SEEMS TO
COMPLICATE MATTERS. WHAT ARE THE ADVANTAGES IN CODEVELOPING DRUG AND DIAGNOSTIC?
ARE THERE DRAWBACKS TO THE DRUG-DIAGNOSTIC CODEVELOPMENT MODEL?
There are drawbacks and interestingly, these arise
precisely from the main task of the diagnostic: to
identify the patient subgroup that responds to the
drug. The standard of clinical trial design used in
the drug-diagnostic codevelopment model is the
enrichment study design. Here only the patients
who test positive with the diagnostic are randomly
assigned to be treated with the tested drug or with a
comparator. In some cases even, all positive patients
are assigned to be treated with the tested drug – no
randomization and no comparator. Approvals from
such trials are based on remarkable response rates
of 50% and higher, where there is little room for
doubt that the use of the diagnostic and the drug
had a positive therapeutic outcome for the patient.
However, the trial design means that the drug is
tested on a small number of patients. In the case of
crizotinib, it was only 255 ALK-positive patients with
non-small cell lung cancer. Safety data about the
drug are therefore limited and the FDA rightfully
mandates in such cases post-approval data to address
the missing information. The benefit is that an
obviously effective drug is available to physicians and
patients as quickly as possible and this works well for
oncology, where most would choose an experimental
or newly approved drug over chemotherapy.
However, the risk to the drug developer is latent.
Another drawback of these trial designs is that by
leaving out the patients who tested negative with the
diagnostic, the trial no longer provides data needed
to determine clinical sensitivity and specificity of
the diagnostic. We know that the diagnostic has
clinical utility — with response rates upwards of
50-60%, the companion diagnostic of crizotinib
4Elsevier RD Solutions
certainly identifies patients who benefit from the drug — but we
know nothing about its sensitivity or specificity. Were patients
who could have benefited from crizotinib not identified? What
patients definitely do not benefit from crizotinib? Those answers
will need to come as more data are collected in continued
post-approval trials. A final drawback worth mentioning is the
endpoint measured in these trials. Response rate is used as a
surrogate of the truly meaningful endpoint: survival. Meta-
analyses for an indication such as non-small cell lung cancer
seems to show that response rate is proportional to survival, but
only long-term data can tell us if a high response rate actually
corresponds to extended survival.
WHEN WE LOOK AT THE LIST OF FDA-APPROVED
COMPANION DIAGNOSTICS, TWO PATTERNS EMERGE.
ONE IS THAT THERE ARE REALLY VERY FEW OF THEM — 20,
AS OF DECEMBER 2014. THE OTHER IS THAT 50% OF THE
APPROVED COMPANION DIAGNOSTICS ARE LINKED TO
HERCEPTIN AND OTHER HER2 TARGETED DRUGS. CAN
YOU EXPLAIN THOSE PATTERNS?
I myself have been a bit surprised by the low number of
companion diagnostics and the rather slow transformation
towards stratified medicine that we have witnessed. However,
I think that this is a changing trend and we will have a “ramp
up” in next years as we learn more about the molecular biology
of diseases and as more drug developers recognize the value of
medicines targeting specific biological subgroups of patients.
Perhaps the second pattern you mention mirrors this change.
Herceptin has been around for a long time, so we understand
the biology quite well. Each new companion diagnostic
associated with the drug and other HER2 targeted drugs
like pertuzumab (Perjeta®, Genentech) and ado-trastuzumab
emtansine (Kadcyla®, Genentech) may emerge for commercial
reasons, but also because of an improved understanding of
the mechanisms of cancer, the introduction of better assay
technologies, or the approval of the drug for new indications,
such as gastric cancer. I think, however, that it is fair to say that
the quantity and diversity of companion diagnostics entering the
clinical setting is and will continue to rise.
5Elsevier RD Solutions
WHAT IS THE FUTURE OF
COMPANION DIAGNOSTICS?
This question brings us back to the
drawbacks I mentioned previously.
The drug-diagnostic codevelopment
model currently operates under the
assumption of “oncogene addiction”
— the postulation that tumors rely on
a single dominant oncogene of growth
and survival so that inhibiting that
oncogene or pathway is sufficient to halt
tumor growth. The result is a “one test,
one drug” construct. There is mounting
evidence that oncogene addiction is, at
best, a transitory condition and tumors
change over time. We already see
resistance development in most patients
on some of these targeted medicines. For
example, ceritinib (Zykadia®, Novartis) is
another lung cancer drug that received
rapid approval and is indicated for
patients who progress on crizotinib, that
is, that have developed resistance to this
drug. As we learn more about the biology
of disease, we can characterize patient
subpopulations in increasing detail and
as a result, drugs will target increasingly
smaller groups of patients. Such precision
in therapy is a welcome prospect of truly
individualized management of disease,
but the construct “one test, one drug” is
not sustainable under those conditions.
From the physician and patient
perspective, this construct would mean an
inordinate number of tests to determine
the right therapy. From a drug developer
perspective, demonstrating drug efficacy
with increasingly smaller patient sample
sizes will become very difficult or
impossible.
In my view, the only solution is a new
multimodal approach. For diagnostics,
I think that will look like multiplexed
panels or arrays that deliver information
on multiple relevant biomarkers. Next-
generation sequencing has the potential
to play a big role here but it will not be the
only platform. Diagnostic information can
also come from proteins, metabolites, or
even functional imaging. The result of this
is that packaging a diagnostic with one
drug may no longer be realistic; instead,
we may need a networked approach
involving multiple drugs. The bottleneck
is the burden on drug developers. We
might know all the right disease-causing
mutations, but we do not have the arsenal
of drugs. I also believe that clinical trial
design will have to evolve. We will need
to step away from the restrictions of
controlled, randomized trials and develop
more flexible and adaptive structures,
including open observational studies,
that enable cooperation and data sharing.
I can envision a knowledge-producing
system that spans multiple clinical trials.
Patient profile and trial outcome data
must meet predetermined minimum
requirements to be compiled in the
system but can then be used to generate
analyses comparing groups between
and among different participating trials.
I do not see this as a purely academic
exercise but one that can also benefit
drug developers. Such a design would
allow them to test their drug on a small
target group but capitalize on the data
in the system as a whole to produce
significant analyses. Furthermore, drug
combinations can more easily be tested
under such a framework.
We need not be naive. Competition will
be an issue for such a model. However,
we are already seeing changes in
mentality that can lead down this path.
Basket trials are an example, where
patients are included in a trial based on
their genetic profile and not on the type
of cancer they have (lung, breast, colon,
etc). This is a retreat from the histological
classification of cancer that has prevailed
for decades but has proven to be less
informative than a classification based
on molecular characteristics. We also
need not be naive about the long path
ahead. We need more drugs. We need a
far better understanding of the molecular
mechanisms underlying cancer and all
other diseases that can benefit from
precision medicine. And while our tools
are getting better and we are getting
smarter, what lies ahead is still small steps
and serial improvement of therapies.
But we are seeing progress. We are
moving forward toward truly personalized
medicine.
“We also need not be naive about
the long path ahead. We need
more drugs. We need a far better
understanding of the molecular
mechanisms underlying cancer and
all other diseases that can benefit
from precision medicine. And while
our tools are getting better and
we are getting smarter, what lies
ahead is still small steps and serial
improvement of therapies. But we
are seeing progress. We are moving
forward toward truly personalized
medicine.”
Visit elsevier.com/rd-solutions
or contact your nearest Elsevier office.
ASIA AND AUSTRALIA
Tel: +65 6349 0222
Email: sginfo@elsevier.com
JAPAN
Tel: +81 3 5561 5034
Email: jpinfo@elsevier.com
KOREA AND TAIWAN
Tel: +82 2 6714 3000
Email: krinfo.corp@elsevier.com
EUROPE, MIDDLE EAST AND AFRICA
Tel: +31 20 485 3767
Email: nlinfo@elsevier.com
NORTH AMERICA, CENTRAL AMERICA AND CANADA
Tel: +1 888 615 4500
Email: usinfo@elsevier.com
SOUTH AMERICA
Tel: +55 21 3970 9300
Email: brinfo@elsevier.com
Copyright© 2015 Elsevier B.V. All rights reserved.
May 2015

More Related Content

What's hot

Medication nonadherence cost and noncompliance in clinical trials
Medication nonadherence cost and noncompliance in clinical trialsMedication nonadherence cost and noncompliance in clinical trials
Medication nonadherence cost and noncompliance in clinical trialsSynegys
 
05 (b)module g doing causality assessment 2_nov05
05 (b)module g doing causality assessment 2_nov0505 (b)module g doing causality assessment 2_nov05
05 (b)module g doing causality assessment 2_nov05Prabir Chatterjee
 
Beyond Traditional Designs in Early Drug Development
Beyond Traditional Designs in Early Drug DevelopmentBeyond Traditional Designs in Early Drug Development
Beyond Traditional Designs in Early Drug DevelopmentMaRS Discovery District
 
Drug risk assessment 23 4-2010
Drug risk assessment 23 4-2010Drug risk assessment 23 4-2010
Drug risk assessment 23 4-2010RobHeerdink
 
Personalised medicine
Personalised medicinePersonalised medicine
Personalised medicineKushal Saha
 
23205042
2320504223205042
23205042radgirl
 
23204998
2320499823204998
23204998radgirl
 
Clinical Proof of Concept (PoC)
Clinical Proof of Concept (PoC)Clinical Proof of Concept (PoC)
Clinical Proof of Concept (PoC)Cytel USA
 
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and  Population PK and Its' ValueUPDATED-Early Phase Drug Developmetn and  Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and Population PK and Its' ValueE. Dennis Bashaw
 
Med adherence and self efficacy
Med adherence and self efficacyMed adherence and self efficacy
Med adherence and self efficacyMarion Sills
 
4 Page ToxProtect Advertorial
4 Page ToxProtect Advertorial4 Page ToxProtect Advertorial
4 Page ToxProtect AdvertorialNate Reed
 
Keynote malone-clinical-relevance-of-ddi-evidence
Keynote malone-clinical-relevance-of-ddi-evidenceKeynote malone-clinical-relevance-of-ddi-evidence
Keynote malone-clinical-relevance-of-ddi-evidenceRichard Boyce, PhD
 
Translation of Orphan DiseaseTrial Design into General Drug Development
Translation of Orphan DiseaseTrial Design into General Drug DevelopmentTranslation of Orphan DiseaseTrial Design into General Drug Development
Translation of Orphan DiseaseTrial Design into General Drug DevelopmentE. Dennis Bashaw
 

What's hot (19)

Medication nonadherence cost and noncompliance in clinical trials
Medication nonadherence cost and noncompliance in clinical trialsMedication nonadherence cost and noncompliance in clinical trials
Medication nonadherence cost and noncompliance in clinical trials
 
Adverse drug reaction causality assessment
Adverse drug reaction causality assessmentAdverse drug reaction causality assessment
Adverse drug reaction causality assessment
 
Bridging the Divide
Bridging the DivideBridging the Divide
Bridging the Divide
 
05 (b)module g doing causality assessment 2_nov05
05 (b)module g doing causality assessment 2_nov0505 (b)module g doing causality assessment 2_nov05
05 (b)module g doing causality assessment 2_nov05
 
Beyond Traditional Designs in Early Drug Development
Beyond Traditional Designs in Early Drug DevelopmentBeyond Traditional Designs in Early Drug Development
Beyond Traditional Designs in Early Drug Development
 
Drug risk assessment 23 4-2010
Drug risk assessment 23 4-2010Drug risk assessment 23 4-2010
Drug risk assessment 23 4-2010
 
Emergency med pharmacist 2017 coralic a. et al
Emergency med pharmacist 2017 coralic a. et alEmergency med pharmacist 2017 coralic a. et al
Emergency med pharmacist 2017 coralic a. et al
 
Personalised medicine
Personalised medicinePersonalised medicine
Personalised medicine
 
Causality Assessment
Causality AssessmentCausality Assessment
Causality Assessment
 
23205042
2320504223205042
23205042
 
23204998
2320499823204998
23204998
 
Clinical Proof of Concept (PoC)
Clinical Proof of Concept (PoC)Clinical Proof of Concept (PoC)
Clinical Proof of Concept (PoC)
 
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and  Population PK and Its' ValueUPDATED-Early Phase Drug Developmetn and  Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
 
10.1007_s11096-016-0258-9
10.1007_s11096-016-0258-910.1007_s11096-016-0258-9
10.1007_s11096-016-0258-9
 
Med adherence and self efficacy
Med adherence and self efficacyMed adherence and self efficacy
Med adherence and self efficacy
 
4 Page ToxProtect Advertorial
4 Page ToxProtect Advertorial4 Page ToxProtect Advertorial
4 Page ToxProtect Advertorial
 
mohammed aiyaz
mohammed aiyazmohammed aiyaz
mohammed aiyaz
 
Keynote malone-clinical-relevance-of-ddi-evidence
Keynote malone-clinical-relevance-of-ddi-evidenceKeynote malone-clinical-relevance-of-ddi-evidence
Keynote malone-clinical-relevance-of-ddi-evidence
 
Translation of Orphan DiseaseTrial Design into General Drug Development
Translation of Orphan DiseaseTrial Design into General Drug DevelopmentTranslation of Orphan DiseaseTrial Design into General Drug Development
Translation of Orphan DiseaseTrial Design into General Drug Development
 

Similar to Elsevier Interview - Dr Jorgensen - May 2015

Accelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapiesAccelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapiesNational Institute of Biologics
 
Genetic Testing Reduces Specialty Drug Spend
Genetic Testing Reduces Specialty Drug SpendGenetic Testing Reduces Specialty Drug Spend
Genetic Testing Reduces Specialty Drug SpendWellDyne
 
InsideView Paving the Path to Precision Medicine (3)
InsideView Paving the Path to Precision Medicine (3)InsideView Paving the Path to Precision Medicine (3)
InsideView Paving the Path to Precision Medicine (3)Saba Anwer, MPH, MBA
 
Five emerging trends
Five emerging trends Five emerging trends
Five emerging trends priya arrora
 
IMPACT STUDY_12NOV2015
IMPACT STUDY_12NOV2015IMPACT STUDY_12NOV2015
IMPACT STUDY_12NOV2015Kim Kersten
 
The paradigm of drug therapy
The paradigm of drug therapyThe paradigm of drug therapy
The paradigm of drug therapyEugene Shorikov
 
BIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUEBIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUEAsmitaGpt
 
PERSONALIZED MEDICINE and customised drug delivery L-1.pptx
PERSONALIZED MEDICINE and customised drug delivery L-1.pptxPERSONALIZED MEDICINE and customised drug delivery L-1.pptx
PERSONALIZED MEDICINE and customised drug delivery L-1.pptxSumant Saini
 
Pcori final writeup
Pcori final writeupPcori final writeup
Pcori final writeuphbocian14
 
Final Research Report - CPOE (Tsourdinis)
Final Research Report - CPOE (Tsourdinis)Final Research Report - CPOE (Tsourdinis)
Final Research Report - CPOE (Tsourdinis)George Tsourdinis
 
Vital Signs Edition #3
Vital Signs   Edition #3Vital Signs   Edition #3
Vital Signs Edition #3ScottJordan
 
Biomarker roles within clinical trials
Biomarker roles within clinical trialsBiomarker roles within clinical trials
Biomarker roles within clinical trialsRay Wright
 

Similar to Elsevier Interview - Dr Jorgensen - May 2015 (20)

Accelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapiesAccelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapies
 
From biomarkers to diagnostics –the road to success
From biomarkers to diagnostics –the road to successFrom biomarkers to diagnostics –the road to success
From biomarkers to diagnostics –the road to success
 
Genetic Testing Reduces Specialty Drug Spend
Genetic Testing Reduces Specialty Drug SpendGenetic Testing Reduces Specialty Drug Spend
Genetic Testing Reduces Specialty Drug Spend
 
Homework
HomeworkHomework
Homework
 
Clinical trials article
Clinical trials articleClinical trials article
Clinical trials article
 
Directed Project
Directed ProjectDirected Project
Directed Project
 
InsideView Paving the Path to Precision Medicine (3)
InsideView Paving the Path to Precision Medicine (3)InsideView Paving the Path to Precision Medicine (3)
InsideView Paving the Path to Precision Medicine (3)
 
OBJECTIVE 3.pptx
OBJECTIVE 3.pptxOBJECTIVE 3.pptx
OBJECTIVE 3.pptx
 
Five emerging trends
Five emerging trends Five emerging trends
Five emerging trends
 
IMPACT STUDY_12NOV2015
IMPACT STUDY_12NOV2015IMPACT STUDY_12NOV2015
IMPACT STUDY_12NOV2015
 
The paradigm of drug therapy
The paradigm of drug therapyThe paradigm of drug therapy
The paradigm of drug therapy
 
CLINICAL TRIALS.pptx
CLINICAL TRIALS.pptxCLINICAL TRIALS.pptx
CLINICAL TRIALS.pptx
 
BIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUEBIOSIMILARS AND ITS FUTUE
BIOSIMILARS AND ITS FUTUE
 
New pattern clinical study
New pattern clinical studyNew pattern clinical study
New pattern clinical study
 
PERSONALIZED MEDICINE and customised drug delivery L-1.pptx
PERSONALIZED MEDICINE and customised drug delivery L-1.pptxPERSONALIZED MEDICINE and customised drug delivery L-1.pptx
PERSONALIZED MEDICINE and customised drug delivery L-1.pptx
 
Pcori final writeup
Pcori final writeupPcori final writeup
Pcori final writeup
 
Precision medicine.ppt
Precision medicine.pptPrecision medicine.ppt
Precision medicine.ppt
 
Final Research Report - CPOE (Tsourdinis)
Final Research Report - CPOE (Tsourdinis)Final Research Report - CPOE (Tsourdinis)
Final Research Report - CPOE (Tsourdinis)
 
Vital Signs Edition #3
Vital Signs   Edition #3Vital Signs   Edition #3
Vital Signs Edition #3
 
Biomarker roles within clinical trials
Biomarker roles within clinical trialsBiomarker roles within clinical trials
Biomarker roles within clinical trials
 

Elsevier Interview - Dr Jorgensen - May 2015

  • 1. Dr. Jan Trøst Jørgensen THE “GATEKEEPER” OF PRECISION MEDICINE EVOLVES. Drug-diagnostic codevelopment can lead to a streamlined approval strategy, but will need to change in the future. PHARMA & LIFE SCIENCES INTERVIEW Director, Dx-Rx Institute
  • 2. 2Elsevier R&D Solutions “...packaging a diagnostic with one drug may no longer be realistic; instead, we may need a networked approach involving multiple drugs.” Dr. Jørgensen has been involved in precision medicine since 1999 and has written several thought-provoking articles on the topic. He has observed this paradigm shift in pharmacotherapy roar through initial hype, wane in interest, and then settle into a steady transformation in the development of new medicines built around the under- standing that diseases and patient populations are heterogeneous. Dr. Jørgensen is a consultant for drug developers and a driving force behind companion diagnostics, which, in his own words, are “a kind of ‘gatekeeper’ in relation to which patients should be treated with the drug in question.” In this interview, Dr. Jørgensen talks about the implications of this gatekeeping role and the future of companion diagnostics. As we learn more about the biology of disease, the construct “one test, one drug” will no longer be sustainable. YOU ARE A STRONG PROPONENT OF THE DRUG-DIAGNOSTIC CODEVELOPMENT MODEL WHERE COLLABORATION BETWEEN DEVELOPERS OF A DRUG AND DEVELOPERS OF A DIAGNOSTIC THAT IDENTIFIES PATIENTS FOR THAT DRUG RESULTS IN A COUPLED, AND AT TIMES EVEN SPEEDIER, APPROVAL OF BOTH. WHAT DOES THIS MODEL LOOK LIKE IN PRACTICE? This co-development model was inspired by the simultaneous FDA approval of the breast cancer medication trastuzumab (Herceptin®, Roche/Genentech) and the immunohistochemical assay for HER2 (HercepTest®, Dako) in 1998. It was also the FDA that first initiated a discussion about drug-diagnostic codevelopment with a 2005 publication where they outlined several important issues for the establishment of routine coupled approvals. Ideally, a drug developer begins working with an assay developer during the discovery and preclinical phases of a drug development program (Figure 1). Starting from a biomarker hypothesis that the assay developer uses to establish a prototype diagnostic, there is a continuous exchange of information between the two parties. The drug developer uses the prototype diagnostic to plan and conduct early clinical trials. The assay developer uses information from clinical efforts to assess the predictive characteristics of the biomarker and establish the appropriate cut-off for patient inclusion, among other things. A continuous and structured dialog between both parties is essential. It should be said, that this early collaboration and iterative exchange is not always reality, however, it is the ideal relationship under which a codevelopment becomes robust and has greater chances of success. Discovery Research Pre-clinical Drug Development Diagnostic Development Parallel Development of Drug and Diagnostic Clinical Phase I Clinical Phase II Clinical Phase III Regulatory Approval Post Approval Phase Biomarker Selection Feasibility Studies Prototype Assay(s) Analytical Validation Clinical Validation Regulatory Approval Clinical Utility Figure 1. The drug-diagnostic codevelopment model builds on a two-way dialog between drug and assay developers. Early collaboration between the two parties coordinates development efforts for drug and diagnostic assay, which results in a strong regulatory approval strategy. Adapted from an original design by Dr. Jan Trøst Jørgensen.
  • 3. 3Elsevier RD Solutions We have to bear in mind that companion diagnostics is in its infancy and codevelopment programs experience all the accompanying growing pains. There is, for example, discord between the regulatory status and consequent requirements of diagnostics in general and the “gatekeeper” role of a companion diagnostic. This is something Daniel Hayes from the University of Michigan, Ann Arbor, and myself have been pointing out for some time. Under our current vision of stratified medicine, where a patient population is subdivided — stratified — into groups based on biological characteristics that are relevant to an individual’s response to a drug, a companion diagnostic is the gatekeeper that determines which patients should be treated with a given drug. As this pivotal node in a patient’s therapy, the diagnostic assay greatly impacts therapeutic outcome and should be subjected to the same level of regulatory requirements as the drug it accompanies. Having said that, the link between drug and diagnostic at the point of therapy extends back to the development and approval process, and this can be very advantageous. Let’s start with the use of the prototype diagnostic to plan and conduct phase I and phase II clinical trials. Information from these stages may feed back to further optimize a compound. Also, the data from these trials are used to establish the cut-off for patient inclusion in phase III trials and, ultimately, clinical use after approval. The right cut-off plays a critical role in the approval strategy for the drug-diagnostic pair. This is a highly complex analysis that can take months to complete and balances relevant biology (hypothesized drug mode of action, disease mechanisms, observed response rates) with the sensitivity and specificity of the diagnostic and several clinical trial design aspects that impact demonstrating the efficacy of the drug. In other words, the codevelopment leads to a better informed and streamlined regulatory approval strategy. Just 5 or 6 years ago, drug developers were hesitant to engage early with diagnostic developers, partly because of the uncertainty of success in drug development. Sometime they would first contact a diagnostic developer after not achieving the targeted efficacy in phase II trials. Now we see companion diagnostics entering the framework much earlier and the result of this front-ended collaboration can be a fast-tracked approval, like that of the lung cancer drug crizotinib (Xalkori®, Pfizer) and its companion diagnostic (VYSIS® ALK Break Apart FISH Probe Kit, Abbott). Thus, uncertainty about a drug should not stop you from working toward a companion diagnostic, because it could rescue your drug! THE REGULATORY APPROVAL OF A NEW DRUG IS A RESOURCE AND TIME-INTENSIVE ENDEAVOR, OFTEN TAKING SEVERAL YEARS TO ACCOMPLISH. ADDING A DIAGNOSTIC ASSAY TO THE PICTURE SEEMS TO COMPLICATE MATTERS. WHAT ARE THE ADVANTAGES IN CODEVELOPING DRUG AND DIAGNOSTIC? ARE THERE DRAWBACKS TO THE DRUG-DIAGNOSTIC CODEVELOPMENT MODEL? There are drawbacks and interestingly, these arise precisely from the main task of the diagnostic: to identify the patient subgroup that responds to the drug. The standard of clinical trial design used in the drug-diagnostic codevelopment model is the enrichment study design. Here only the patients who test positive with the diagnostic are randomly assigned to be treated with the tested drug or with a comparator. In some cases even, all positive patients are assigned to be treated with the tested drug – no randomization and no comparator. Approvals from such trials are based on remarkable response rates of 50% and higher, where there is little room for doubt that the use of the diagnostic and the drug had a positive therapeutic outcome for the patient. However, the trial design means that the drug is tested on a small number of patients. In the case of crizotinib, it was only 255 ALK-positive patients with non-small cell lung cancer. Safety data about the drug are therefore limited and the FDA rightfully mandates in such cases post-approval data to address the missing information. The benefit is that an obviously effective drug is available to physicians and patients as quickly as possible and this works well for oncology, where most would choose an experimental or newly approved drug over chemotherapy. However, the risk to the drug developer is latent. Another drawback of these trial designs is that by leaving out the patients who tested negative with the diagnostic, the trial no longer provides data needed to determine clinical sensitivity and specificity of the diagnostic. We know that the diagnostic has clinical utility — with response rates upwards of 50-60%, the companion diagnostic of crizotinib
  • 4. 4Elsevier RD Solutions certainly identifies patients who benefit from the drug — but we know nothing about its sensitivity or specificity. Were patients who could have benefited from crizotinib not identified? What patients definitely do not benefit from crizotinib? Those answers will need to come as more data are collected in continued post-approval trials. A final drawback worth mentioning is the endpoint measured in these trials. Response rate is used as a surrogate of the truly meaningful endpoint: survival. Meta- analyses for an indication such as non-small cell lung cancer seems to show that response rate is proportional to survival, but only long-term data can tell us if a high response rate actually corresponds to extended survival. WHEN WE LOOK AT THE LIST OF FDA-APPROVED COMPANION DIAGNOSTICS, TWO PATTERNS EMERGE. ONE IS THAT THERE ARE REALLY VERY FEW OF THEM — 20, AS OF DECEMBER 2014. THE OTHER IS THAT 50% OF THE APPROVED COMPANION DIAGNOSTICS ARE LINKED TO HERCEPTIN AND OTHER HER2 TARGETED DRUGS. CAN YOU EXPLAIN THOSE PATTERNS? I myself have been a bit surprised by the low number of companion diagnostics and the rather slow transformation towards stratified medicine that we have witnessed. However, I think that this is a changing trend and we will have a “ramp up” in next years as we learn more about the molecular biology of diseases and as more drug developers recognize the value of medicines targeting specific biological subgroups of patients. Perhaps the second pattern you mention mirrors this change. Herceptin has been around for a long time, so we understand the biology quite well. Each new companion diagnostic associated with the drug and other HER2 targeted drugs like pertuzumab (Perjeta®, Genentech) and ado-trastuzumab emtansine (Kadcyla®, Genentech) may emerge for commercial reasons, but also because of an improved understanding of the mechanisms of cancer, the introduction of better assay technologies, or the approval of the drug for new indications, such as gastric cancer. I think, however, that it is fair to say that the quantity and diversity of companion diagnostics entering the clinical setting is and will continue to rise.
  • 5. 5Elsevier RD Solutions WHAT IS THE FUTURE OF COMPANION DIAGNOSTICS? This question brings us back to the drawbacks I mentioned previously. The drug-diagnostic codevelopment model currently operates under the assumption of “oncogene addiction” — the postulation that tumors rely on a single dominant oncogene of growth and survival so that inhibiting that oncogene or pathway is sufficient to halt tumor growth. The result is a “one test, one drug” construct. There is mounting evidence that oncogene addiction is, at best, a transitory condition and tumors change over time. We already see resistance development in most patients on some of these targeted medicines. For example, ceritinib (Zykadia®, Novartis) is another lung cancer drug that received rapid approval and is indicated for patients who progress on crizotinib, that is, that have developed resistance to this drug. As we learn more about the biology of disease, we can characterize patient subpopulations in increasing detail and as a result, drugs will target increasingly smaller groups of patients. Such precision in therapy is a welcome prospect of truly individualized management of disease, but the construct “one test, one drug” is not sustainable under those conditions. From the physician and patient perspective, this construct would mean an inordinate number of tests to determine the right therapy. From a drug developer perspective, demonstrating drug efficacy with increasingly smaller patient sample sizes will become very difficult or impossible. In my view, the only solution is a new multimodal approach. For diagnostics, I think that will look like multiplexed panels or arrays that deliver information on multiple relevant biomarkers. Next- generation sequencing has the potential to play a big role here but it will not be the only platform. Diagnostic information can also come from proteins, metabolites, or even functional imaging. The result of this is that packaging a diagnostic with one drug may no longer be realistic; instead, we may need a networked approach involving multiple drugs. The bottleneck is the burden on drug developers. We might know all the right disease-causing mutations, but we do not have the arsenal of drugs. I also believe that clinical trial design will have to evolve. We will need to step away from the restrictions of controlled, randomized trials and develop more flexible and adaptive structures, including open observational studies, that enable cooperation and data sharing. I can envision a knowledge-producing system that spans multiple clinical trials. Patient profile and trial outcome data must meet predetermined minimum requirements to be compiled in the system but can then be used to generate analyses comparing groups between and among different participating trials. I do not see this as a purely academic exercise but one that can also benefit drug developers. Such a design would allow them to test their drug on a small target group but capitalize on the data in the system as a whole to produce significant analyses. Furthermore, drug combinations can more easily be tested under such a framework. We need not be naive. Competition will be an issue for such a model. However, we are already seeing changes in mentality that can lead down this path. Basket trials are an example, where patients are included in a trial based on their genetic profile and not on the type of cancer they have (lung, breast, colon, etc). This is a retreat from the histological classification of cancer that has prevailed for decades but has proven to be less informative than a classification based on molecular characteristics. We also need not be naive about the long path ahead. We need more drugs. We need a far better understanding of the molecular mechanisms underlying cancer and all other diseases that can benefit from precision medicine. And while our tools are getting better and we are getting smarter, what lies ahead is still small steps and serial improvement of therapies. But we are seeing progress. We are moving forward toward truly personalized medicine. “We also need not be naive about the long path ahead. We need more drugs. We need a far better understanding of the molecular mechanisms underlying cancer and all other diseases that can benefit from precision medicine. And while our tools are getting better and we are getting smarter, what lies ahead is still small steps and serial improvement of therapies. But we are seeing progress. We are moving forward toward truly personalized medicine.”
  • 6. Visit elsevier.com/rd-solutions or contact your nearest Elsevier office. ASIA AND AUSTRALIA Tel: +65 6349 0222 Email: sginfo@elsevier.com JAPAN Tel: +81 3 5561 5034 Email: jpinfo@elsevier.com KOREA AND TAIWAN Tel: +82 2 6714 3000 Email: krinfo.corp@elsevier.com EUROPE, MIDDLE EAST AND AFRICA Tel: +31 20 485 3767 Email: nlinfo@elsevier.com NORTH AMERICA, CENTRAL AMERICA AND CANADA Tel: +1 888 615 4500 Email: usinfo@elsevier.com SOUTH AMERICA Tel: +55 21 3970 9300 Email: brinfo@elsevier.com Copyright© 2015 Elsevier B.V. All rights reserved. May 2015