Skip to Main Content

YSPH Biostatistics Seminar: "Innovations in Immune-Oncology Early-Phase Trial Designs: Theory, Practice and Next Steps"

November 08, 2021
  • 00:00<v Wei>Okay, hello everyone.</v>
  • 00:02Today, we are very fortunate to have Dr. Codruta Chiuzan
  • 00:11as our speaker.
  • 00:14Dr. Chiuzan is Associated Professor,
  • 00:17Institute of Health System Science
  • 00:20at Northwell Health New York.
  • 00:23So before that, she was an Assistant Professor
  • 00:26in the Department of Biostatistics
  • 00:29at Mailman School of Public Health Columbia University.
  • 00:33In her research area focus on earning phase
  • 00:37clinical trial designs and an average aging real-world
  • 00:40evidence to prove all the cons and increased diversity
  • 00:44of population in clinical trials.
  • 00:47Now, she is in receipt of Junior Faculty Research Award
  • 00:52and the Columbia Public Health Innovation Award
  • 00:55from the Mailman School of Public Health.
  • 00:58So, Dr. Chiuzan, has a very strong record or mentoring,
  • 01:03both master's students, PhD students and clinical fatal.
  • 01:08She is an active committee member
  • 01:10of JSM, Diversity Mentoring Program
  • 01:13and she had held leadership positions
  • 01:15as the President of the American Statistical Association
  • 01:19and New York City Metropolitan Area
  • 01:22and she as the Chair over the Student Scholars Committee
  • 01:26at the Society of Clinical Trials.
  • 01:29So, welcome Dr. Chiuzan and time is yours.
  • 01:35<v ->Thank you so much Wei, for the invitation,</v>
  • 01:39it's a pleasure, I'm going to share my...
  • 01:47Hello.
  • 01:50Hello, I hear some echo in the background.
  • 01:56<v Student>Is anybody (indistinct).</v>
  • 02:10<v ->The host, can you disable the screen sharing</v>
  • 02:13so I can share the slides?
  • 02:25Oh, perfect.
  • 02:40Okay, can everybody see the screen, the full screen?
  • 02:57Okay.
  • 03:01All right, so it's a pleasure to be with you
  • 03:07even virtually and I'm glad to see so many people in person
  • 03:13with academic semester ongoing.
  • 03:20Today I'm going to talk about one area of my research
  • 03:25and that is early phase designs for immunotherapies
  • 03:33or cancer immunotherapies and I will take you
  • 03:39through a journey, through a story by giving some examples
  • 03:43and explanation of what are these cancer immunotherapies,
  • 03:47what are the promises, what are the challenges
  • 03:50and how do they actually reflect in the early phase designs?
  • 03:57Then I will talk about current models
  • 04:01and a model that we developed on has been implemented
  • 04:06and has been implemented in an R package and the Shiny app
  • 04:12and I will conclude with a practical demonstration.
  • 04:18Please, if anybody has any questions at any point,
  • 04:24please feel free to raise a hand or just ask,
  • 04:27I like us to have an interactive session
  • 04:32and to have a continuous dialogue if you're able please.
  • 04:39So what is immunotherapy?
  • 04:42The New York times called it a long awaited reality
  • 04:45because immunotherapy has been developed
  • 04:50since the early 1900s, actually by a New York surgeon
  • 04:56that saw that in cancer patients that develop flu,
  • 05:03had a better anti-cancer response.
  • 05:07So immunotherapy works on a different paradigm
  • 05:11compared to cytotoxic agents and by cytotoxic agents,
  • 05:15I mean, chemotherapy or radiations.
  • 05:19So immunotherapy boosts or leverages the body's own immune
  • 05:22system to fight cancer, to recognize it, to attack it
  • 05:26and ultimately to kill the cancer cells.
  • 05:29The three Rs of cancer immunotherapies
  • 05:32are reverse tolerance, rejuvenate the immune system
  • 05:39and restore the internal environment homeostasis.
  • 05:46So, you'll probably hear more and more updates
  • 05:51and FDA approvals for cancer immunotherapies.
  • 05:55Between 2017 and 2020, over 65% increase has been seen
  • 06:01in the number of immunotherapies and these immunotherapies,
  • 06:05most of them have been approved
  • 06:06for immune checkpoint inhibitors, Ipilimumab, Nivolumab
  • 06:12and Pembrolizumab that works by incubator thing,
  • 06:17the relationship, the association between the PD-1
  • 06:20and the PD-L1 receptors, but the largest growth
  • 06:27has been seen for cell therapies.
  • 06:31And what are these cell therapies?
  • 06:34The most frequent and the most study one is called T-cells,
  • 06:40so far, which will be approvals.
  • 06:42So that these old therapies use the T-cells in the body
  • 06:48to fight cancer and then you do that by first,
  • 06:54taking blood from the patient, isolating the T-cells
  • 06:58in the lab in the Petri dish and genetically modify
  • 07:02these T-cells to display a specific receptor
  • 07:07that then is introduced and after that when the cells
  • 07:14are being introduced into the body, this T-cell receptor
  • 07:17will bind to specific antigen present on the cancer cells
  • 07:22and trigger an anti-tumor reaction.
  • 07:26So these are the T-cells, these are the cell therapies
  • 07:33that are being studied and they're very promising
  • 07:36in terms of prolonging overall survival
  • 07:39and also in lowering the toxicity,
  • 07:43killing cancer without killing the patient.
  • 07:48Last year update from the Cancer Research Institute
  • 07:52has shown that, as I said, that the most promising field
  • 07:57in cancer therapies are,
  • 07:59has been seen in this T-cell therapies,
  • 08:02most of them for a solid cancer, nonsmall,
  • 08:07renal cancer, colorectal cancer,
  • 08:10but it's moving into the non-solid cancer as well.
  • 08:17So, Hype versus Hope, I entitled this slide,
  • 08:22immunotherapy, is it the holy grail
  • 08:27is the answer to all cancer therapies, yes or no,
  • 08:32because of course it comes with some challenges.
  • 08:36Some of the challenges are immunotherapy sometimes
  • 08:41can trigger delayed responses,
  • 08:44meaning that the treatment has to continue
  • 08:46even if initial response has not been seen,
  • 08:50there's been cases of hyper-progression
  • 08:53where the cancer tumor seen this rapid growth
  • 08:57in the early stages and that can be the problematic
  • 09:04on diminished overall survival.
  • 09:07And most importantly, we're not really sure exactly
  • 09:11what to measure and how to incorporate end points
  • 09:17into all phases of drug development from early phase,
  • 09:22phase one and two, where we're looking at identifying
  • 09:24the optimal dose to later phases.
  • 09:29So, because it's still under development,
  • 09:33there is a lack of biomarkers to predict the responders
  • 09:37versus not the responders and the difficult to correlate
  • 09:41these immunological biomarkers with outcomes,
  • 09:44clinical outcomes, overall survival response progression,
  • 09:48free survival compared to cytotoxic agents, chemotherapy,
  • 09:52immunotherapies have different toxicity profiles,
  • 09:56meaning they can have lower toxicity
  • 09:59and also different grades and different profiles,
  • 10:06is called by, also called as immune-related adverse event.
  • 10:13So if you're familiar with drug development phases,
  • 10:16as you know that usually it will start with early phase.
  • 10:20Phase one, identifying the maximum tolerated dose
  • 10:24to be carried forward then for establishing efficacy
  • 10:28and in later phases.
  • 10:32In the old paradigm, so the objective was to find the MTD
  • 10:37and the MTD was mainly based on toxicity as binary,
  • 10:42yes or no DLT, the patient has after receiving treatment,
  • 10:48we quantify the number of those limiting toxicities,
  • 10:51unacceptable toxicity within a certain interval.
  • 10:55However, the new immunotherapies have,
  • 10:59as I mentioned before,
  • 11:00they have different toxicity profiles,
  • 11:02so this old paradigm of finding the MTD,
  • 11:07does longer stint, there are a lot of trials
  • 11:10where the dose escalation moved quickly
  • 11:14to the maximum dose level and MTD.
  • 11:17There were no DLTs, the MTD was not identified
  • 11:21and most importantly, toxicity and efficacy
  • 11:26might not necessarily be those dependents.
  • 11:28So you might be able to find a safe dose,
  • 11:31but that might not necessarily be the most promising one
  • 11:36in terms of efficacy.
  • 11:37In many cases, we actually see this plateau trend
  • 11:41where after a certain level,
  • 11:43the efficacy levels out plateaus
  • 11:47and we don't see any effect.
  • 11:52Another challenge, so in this context of different toxicity,
  • 11:58is different levels of toxicity incorporation of efficacy
  • 12:00into the dose finding process, we need to reconsider
  • 12:06the definition and think of more in terms of identifying
  • 12:11the optimal biological dose versus the MTD,
  • 12:15a dose that is acceptable in terms of toxicity,
  • 12:18but also this place, a good efficacy profile.
  • 12:25So in terms of methodology, again in early-phase,
  • 12:33research has been dominated in the past decades
  • 12:38by algorithmic designs by algorithmic,
  • 12:41I mean the three plus three,
  • 12:42which is definitely not preferred by,
  • 12:49and actually strongly disapproved by statisticians
  • 12:55and even until 2014, when we did the last review
  • 13:02of early-phase methodology, we saw that over 90%
  • 13:05of this trials have implemented a rule-based design.
  • 13:10Rule-based working only on toxicity with absolutely
  • 13:14no statistical background.
  • 13:20From 2012, we saw more than 60% of the trials
  • 13:25that the tested targeted or immunotherapies
  • 13:29and only 7.6 actually used a model-based design.
  • 13:34So what do I mean by model-based design?
  • 13:36Well, we criticize in three plus three,
  • 13:39but are there alternatives actually several.
  • 13:45One alternative that addresses the matter
  • 13:50of late onset toxicities that is usually seen
  • 13:54in immunotherapies, meaning you see DLTs on toxicity
  • 13:58outside of DLT window, that is usually 28 days
  • 14:04so longer toxicities, for that we have the time to event
  • 14:09continual reassessment method that was proposed
  • 14:12by Jenga Chapelle in 2000.
  • 14:16The problem with multiple toxicities
  • 14:19across different varying grades and moving away
  • 14:22from the binary DLT has been tackled by Ezzalfani and others
  • 14:27by using, by incorporating these types,
  • 14:30different toxicity types and different grades
  • 14:33into the total toxicity score,
  • 14:35which is a quasi continuous measure.
  • 14:40As I mentioned for immunotherapies,
  • 14:43it makes more sense to incorporate both toxicity
  • 14:46and efficacy and for that we have models that look at,
  • 14:53that incorporate both toxicity and efficacy
  • 14:56and these are the F-stocks designs method
  • 15:00or the bivariate continual reassessment method.
  • 15:05And more recently, other measures have been looked
  • 15:10at in immunotherapies and these are the PK,
  • 15:14the pharmacokinetics or the pharmacodynamics
  • 15:17and these have been incorporated by, for example,
  • 15:20Ursino, in a patient design proposed in 2017.
  • 15:25So this is to present the status quo
  • 15:29of what's being proposed out there, what we are suggesting
  • 15:33is also a design that is specific
  • 15:38or for immunotherapy trials and this was published in 2018
  • 15:44and since then we have added a different measure
  • 15:47of toxicity, we have implemented it into an R package
  • 15:51that is on a available on cram, iAdapt
  • 15:55and also can be tried using charmia.
  • 15:59So this design for immunotherapies uses both toxicity
  • 16:04and efficacy to identify the optimal dose.
  • 16:09Optimal dose meaning unacceptable dose
  • 16:11with promising efficacy profile.
  • 16:16The design is unique in the sense it can incorporate
  • 16:19both binary or quasi continuous toxicity scores,
  • 16:23and it's looking at the continuous efficacy outcomes.
  • 16:26Most of the designs that I mentioned before are using
  • 16:29either binary or ordinal efficacy.
  • 16:32In this one we're looking at continuous outcomes,
  • 16:35such as T-cell persistence at followup compared to baseline
  • 16:39why the cell persistence, as I mentioned before,
  • 16:43well, about this engineered T-cells
  • 16:46when they are being put into the body,
  • 16:48they maintain the steer soul store,
  • 16:51then the genetic information and the trigger
  • 16:58and tumor response and it's been shown,
  • 17:02there's some studies shown that the number of T-cells
  • 17:06that are still present, still survive in the blood
  • 17:10at one or two months after being reinfused tends to predict
  • 17:17response on the overall survival on the long-term.
  • 17:22The design has, does not impose any monotonicity assumption
  • 17:26in terms of those efficacy relationship and does not account
  • 17:30for dependence between toxicity and efficacy.
  • 17:34So now let's take a look at the two,
  • 17:37the difference between incorporating toxicity only
  • 17:40and looking at efficacy also.
  • 17:43So the cartoon on the left shows the dose toxicity
  • 17:47relationship or five dose level.
  • 17:50So in this graph, let's say we have five dose levels
  • 17:53and we have a threshold of unacceptable toxicity set at 40%.
  • 17:58So based on this graph, we have about four dose levels
  • 18:04that are below the threshold, one dose level that is above.
  • 18:11So if we have 40% toxicity threshold, dose number four
  • 18:18would be identified as the MTD, the maximum tolerated dose.
  • 18:22However, if we are to look also at efficacy
  • 18:27and in this case, the dose efficacy has this umbrella,
  • 18:30this non-monitoring trend.
  • 18:32We will see that by looking at the MTD,
  • 18:35we would totally miss the optimal dose
  • 18:39because dose number four has actually a lower efficacy
  • 18:46as compared to dose number three.
  • 18:49So this is to pretty much justify the need to incorporate
  • 18:53both toxicity and efficacy into the dose finding process.
  • 18:59So the design has two stages.
  • 19:01In stage one, we're establishing the safety profile
  • 19:05at each dose, after we establish the safety profile,
  • 19:09the acceptable doses are carried to stage number two,
  • 19:13where we using efficacy driven randomization
  • 19:15to allocate patients to acceptable doses
  • 19:19that emphasis towards more promising efficacious ones.
  • 19:25So, now let's take a look at stage one,
  • 19:27establishing the safety profile.
  • 19:29We have a number of pre-specified dose levels
  • 19:33and we start by defining the set of hypothesis,
  • 19:37where hypothesis one represents the unacceptable DLT rate
  • 19:42and hypothesis two, represent unacceptable DLT rate.
  • 19:46DLT, meaning the Dose Limiting Toxicity.
  • 19:53So the quantification of this evidence
  • 19:59in favor of hypothesis one or hypothesis two,
  • 20:04is done by the likelihood ratio V, the evidential paradigm.
  • 20:10So to give you a little bit of a background,
  • 20:15in statistics there pretty much three school of thoughts,
  • 20:18we have the frequencies approach based on Pearson,
  • 20:22you'll have the patient school of thought,
  • 20:24and then you have the evidential paradigm.
  • 20:27The evidential paradigm and the frequent tests
  • 20:30are somehow similar, but the difference between the two
  • 20:35is the evidential paradigm based on the law of likelihood
  • 20:39the couples, the strength of evidence from uncertainty.
  • 20:43So the strength of the evidence is quantified
  • 20:45by the likelihood ratio and our certainty
  • 20:48is quantified by the probability of misleading evidence
  • 20:54and the probability of observing weak or strong evidence
  • 20:59in favor of the other two.
  • 21:03Yeah, in comparison the frequent is the approach,
  • 21:05that's not the couple, the strength of evidence
  • 21:09and from uncertainty.
  • 21:12So evidential paradigm used to establish acceptability
  • 21:20in stage number one.
  • 21:21And how do we do that?
  • 21:22Let's say we have a certain number of levels, each show,
  • 21:27and we treat cohorts of size M patients
  • 21:32to each of these dose levels based on toxicity information,
  • 21:38we calculate the likelihood ratio and evaluate evidence
  • 21:44as one of the three, either we have strong evidence
  • 21:48in favor of hypothesis two,
  • 21:51declaring that the dose is acceptable.
  • 21:54Either we have strong evidence in favor of H1,
  • 21:57it's unacceptable or we conclude the weak evidence
  • 22:02that doesn't support either of the hypothesis,
  • 22:05the likelihood ratio is compared to a threshold, okay?
  • 22:10So how do we, let's take a look at an example
  • 22:13to see how we set the hypothesis
  • 22:15and how we started this threshold, okay?
  • 22:18So let's say that we have hypothesis one 40%,
  • 22:23this is unacceptable, the DLT rate toxic
  • 22:27and hypothesis two 15%, that is an acceptable DLT rate
  • 22:34and we want, we evaluate each dose
  • 22:37based on these two hypothesis.
  • 22:40And for that one, in this case, we use a k threshold
  • 22:46equal to two, so there's been a lot of literature
  • 22:51written on this evidential paradigm and the k thresholds
  • 22:56can vary, we can take values from two, four, eight
  • 23:02all the way to 32, depending on the sample size,
  • 23:05the bigger the sample size, the bigger the k thresholds.
  • 23:11Because in phase one, we tend to deal with limited
  • 23:15sample sizes, 30 maybe all the way to 50 number of patients,
  • 23:22of k threshold or two or four seems to be sufficient
  • 23:28to be able to quantify the strength of evidence.
  • 23:31So I teach dose levels based on cohorts of three patients,
  • 23:36we compare the likelihood ratio and if the likelihood ratio
  • 23:40is greater than one over k in this case two,
  • 23:44we decide that the dose is acceptable and safe
  • 23:46and it will be carried forward to station number two.
  • 23:50Otherwise, the dose is considered unacceptably toxic
  • 23:55and it's being discarded and will not be considered
  • 23:59for further evaluation.
  • 24:01So in this case, let's say that we have two or more
  • 24:06the maximum doses in stage one, we continue to stage two
  • 24:11to employ an adaptive randomization.
  • 24:16So in stage two, we use a linear model
  • 24:19to calculate the randomization probabilities
  • 24:21based on efficacy and in this case we use indicator variable
  • 24:26for each dose level and Y represents the continuous
  • 24:32immunological response and as I mentioned before
  • 24:36in our application, that response is a T-cell persistence
  • 24:41at one month after infusion.
  • 24:44So based on the estimated Ys,
  • 24:48we calculate the randomization probability for each dose
  • 24:53and allocate patients sequentially based on this path.
  • 24:58So how does this look, let's say again,
  • 25:01that we have for dose levels and three patients treated
  • 25:05at each dose level, we measure the T-cell persistence
  • 25:10for all patients within the cohort
  • 25:12and we feed the linear model
  • 25:16to generate the estimated efficiencies.
  • 25:21Based on the estimated efficiencies,
  • 25:23we calculate the randomization probabilities.
  • 25:27So for each dose, in this case,
  • 25:30the randomization probability for dose one is 5%,
  • 25:34the highest is for dose number 49.
  • 25:37So the next patient that will be allocated,
  • 25:43that will be randomized, will probably be randomized
  • 25:48to dose number four,
  • 25:49because this one has the highest randomization probability,
  • 25:54and the process continues in stage two
  • 25:56and feel you have reached the maximum sample size
  • 26:00that you've specified for the trial.
  • 26:05So how did we evaluate the model behavior?
  • 26:09Well, we looked at two different sample sizes,
  • 26:1225 and 50 patients in total for the trial.
  • 26:17The number of levels varied from three to five,
  • 26:20we don't recommend using this design
  • 26:25for less than three of dose levels is just not enough
  • 26:28and the design that you don't gain anything by using it
  • 26:33if you have less than three.
  • 26:36We use the 5,000 simulations for each scenario
  • 26:41and in terms of establishing the operating characteristics,
  • 26:46we quantified two things.
  • 26:48One, we looked at present those allocation
  • 26:52and we looked at estimation of efficacy outcomes,
  • 26:55because that is the main goal actually, after implementing.
  • 26:59you're looking at both toxicity and efficacy
  • 27:03in determining the optimal dose
  • 27:05with the goal of allocating more patients,
  • 27:08skewing the allocation to a dose that is acceptably safe
  • 27:13and has promising efficacy,
  • 27:17in this case has a higher percentage of these helper system.
  • 27:24So this is our combination of efficacy and toxicity scenario
  • 27:29in panel A you see that we have five dose level
  • 27:35on the x-axis and on the y we have the T-cell persistence
  • 27:39as a functional skills
  • 27:41and the scenarios vary from completely flat to monotonic
  • 27:47increasing to non-monotonic and also to plateau,
  • 27:53which is scenario number three.
  • 27:55As I mentioned, this is a pretty frequent scenario
  • 27:59at a certain dose level, the efficacy, that's not,
  • 28:05we don't see any big increases in the efficacy.
  • 28:08In terms of toxicity, again, different flat trends
  • 28:15on the steeper dose toxicities scenario.
  • 28:22In terms of beta stimulation, toxicity was stimulated
  • 28:26from a Bernoulli distribution, persistence was simulated
  • 28:31from a beta-binomial and for variance,
  • 28:35the variance was assumed to be a constant,
  • 28:40but we then read the values of small and large,
  • 28:44and to give you an idea of what means
  • 28:45a large variance at 1%, that is equivalent to about 20%
  • 28:50deviation from the mean T-cell persistence
  • 28:55toxicity and efficacy are modeled independently.
  • 29:01So some results, the paper on had the has several scenarios,
  • 29:08but just to illustrate, this is for a total sample size
  • 29:14of 25 patients both in stage one and in stage two
  • 29:18and the design was compared to MTPI,
  • 29:21which is the Modified Toxicity Probability Interval.
  • 29:25And we wanted to compare and this is one of the operating
  • 29:29correctors, 6% patient allocation.
  • 29:34How does the design allocate patients based on toxicity
  • 29:38and efficacy?
  • 29:39So in this case, that toxicity we see is toxicity three,
  • 29:46we have five, dose levels, the three dose levels
  • 29:51have toxicities lower than 15%.
  • 29:56Dose number four has a toxicity between 15 and 40%,
  • 30:02and dose number five pretty much
  • 30:05is at the maximum DLT threshold,
  • 30:08'cause remember that we had two hypothesis,
  • 30:1115% acceptable toxicity and 40%, so dose one, two, three
  • 30:15are considered acceptable.
  • 30:17Dose four is within the interval,
  • 30:1915, 40 and 40% as shown in red is considered a toxic dose.
  • 30:28So this is the same toxicity
  • 30:31for different efficacy scenarios increasing,
  • 30:35this is non-monotonic this umbrella plateau,
  • 30:41this umbrella trend,
  • 30:42this efficacy three is the plateau trend
  • 30:46and the efficacy four is constant efficacy across all doses.
  • 30:54So what we noticed that the design does a good job
  • 30:59allocating most of the patients
  • 31:01to a dose that is considered safe
  • 31:04and also has the optimal efficacy.
  • 31:10So this would be dose number three in panel A,
  • 31:15again, dose number three in panel B and similar for C
  • 31:22and so ultimately we allocate most of the patients
  • 31:29to this optimal level.
  • 31:35Now let me, I would like to show you an illustration
  • 31:40of how can we use this design,
  • 31:44its implementation in the R package and in the Shiny app,
  • 31:50because the package has two purposes.
  • 31:53One, is to run simulation, to observe to quantify
  • 31:57the operating correctly sticks for different dose,
  • 32:01toxicity dose efficacy scenarios and another benefit
  • 32:07is that you can implement it to actually allocate,
  • 32:11to run the trial, to allocate the next patient
  • 32:17to the optimal dose.
  • 32:18So simulation and implementation and the scenario
  • 32:23that I will discuss next is inspired from a real study
  • 32:29that we worked on when I was at the Cancer Center
  • 32:35at Columbia and this was a phase-one trial
  • 32:38evaluating modified autologous T-cells
  • 32:41this genetically modified T-cells in patients
  • 32:46where the recurrent solid tumors.
  • 32:48So if you think that all these designs
  • 32:51are usually theoretical or just great statistical proposals,
  • 32:55actually this one had an implementation
  • 33:03and had a real setting.
  • 33:05So the initial design that was proposed
  • 33:12was of course, an role-based design derivation,
  • 33:16this was a two-by-two with up to five dose levels,
  • 33:19and I wanna bring your attention to the dose levels.
  • 33:23So in immunotherapy, especially in this T-cell therapies,
  • 33:28the dose levels are not quantities of dose age
  • 33:34of the medication are not milligrams,
  • 33:36but they are actually the number of T-cells
  • 33:41that are being infused back into the body.
  • 33:44So in this case the dose levels vary from 50 to 10
  • 33:53to the six to 500 and 10 the six viable T-cells,
  • 33:58so millions of cells.
  • 34:05And we wanted to explore what is the optimal dose
  • 34:13for this regimen.
  • 34:16So, this is a snapshot of the Shiny app,
  • 34:26we redesigned the trial to incorporate both toxicity
  • 34:29and continuous efficacy and in this case continue,
  • 34:33this T-cell persistence was a reasonable biomarker,
  • 34:38we looked at five dose levels, we had in simulations,
  • 34:43you have to specify what are the toxicity to toxicity rates
  • 34:47and what are the T-cell persistence levels.
  • 34:52The true toxicity rates varied from five to 40%,
  • 34:55the T-cell persistence varied from 15 to 40%.
  • 35:00We didn't see that we're going to see more than 40%
  • 35:03persistence followup and we looked at the total sample size
  • 35:08of 30 patients for the trial, this was feasible
  • 35:14and practical.
  • 35:16So the setup it's very simple,
  • 35:19you just put the number of dose levels,
  • 35:21you specify the toxicities, you specify the mean efficacy,
  • 35:25the variance for the efficacy, we chose 1%,
  • 35:29but this can take different values,
  • 35:35then of course, this is a dialogue
  • 35:37that you have with your clinical investigator,
  • 35:42hopefully with data supported from previous studies
  • 35:45and what is considered to be to very these parameters.
  • 35:50Then for stage one, you have to specify the two hypothesis,
  • 35:54acceptable and unacceptable DLT.
  • 35:58For this one, we set it at 15 and 40%,
  • 36:02the likelihood ratio was set at two
  • 36:05because of the sample size of 30 with k equals to two
  • 36:12is reasonable and three cohorts, three patients per cohort,
  • 36:17meaning each of the five dose levels we have three patients
  • 36:22allocated each.
  • 36:24Total sample size of 30 on the stopping rule,
  • 36:28stopping rule meaning if none of the doses
  • 36:32are considered acceptable in stage one,
  • 36:35we're going to allocate up to nine patients
  • 36:40at the first dose to further establish toxicity,
  • 36:43and this can be changed to six or other number.
  • 36:52So these are, how did the scenario looks?
  • 36:56This is the graphs actually generated by the app
  • 37:01toxicity and efficacy as a function of dose level,
  • 37:05and now I'd like to ask you, ask for your participation,
  • 37:12looking at these two scenarios,
  • 37:14what do you think would be an optimal dose level,
  • 37:19the recommended to be studied in phase-two or later?
  • 37:35Okay, maybe we need some hint,
  • 37:39so we want an acceptable dose.
  • 37:44<v ->[] Three.</v>
  • 37:46<v ->Dose number three, dose number three,</v>
  • 37:49because dose number three is way outside
  • 37:55of the acceptability range and also dose number three
  • 38:00tends to have a good efficacy after dose number three
  • 38:06we don't see any improvement in terms of efficacy.
  • 38:11Dose number four is between the 15 and 40%
  • 38:14then dose number five was probably toxic.
  • 38:18So, dose number three is the optimal dose
  • 38:21and what we would like to see,
  • 38:26is most patients being allocated at this level.
  • 38:29So in simulations, this is simulations for stage one,
  • 38:37where based on observed the DLTs,
  • 38:40we calculate the likelihood ratio and mark the doses
  • 38:49as being acceptable or unacceptable.
  • 38:53So in this case, based on the simulations, we see dose one,
  • 38:57two, three and four are considered acceptably safe
  • 39:03and they will be carried forward to stage number two,
  • 39:07to be considered for that different organization,
  • 39:10dose number five will be discarded
  • 39:13and will not be used in stage two.
  • 39:16And why is that?
  • 39:17Because the likelihood ratio is less than 0.05.
  • 39:29Now these are the simulations for a stage number two,
  • 39:33so in the first part, we had five dose levels,
  • 39:37three patients each, so that's a total of 15 patients
  • 39:40starting with patient number 16, we moved to stage two
  • 39:45and we do this adaptive randomization
  • 39:47until we reach the maximum sample size of 30.
  • 39:51So the app actually gives you simulations and allocations
  • 39:58for all the patients from 16 to 30 dose assignments
  • 40:02and efficacy outcome and this gives you a graph
  • 40:07of the estimated efficacy and the medians
  • 40:13and inter quartile ranges.
  • 40:17So we repeated this a hundred times, you can repeat it more
  • 40:211,000, 5,000 in terms of allocation based on the setting,
  • 40:29based on the parameters, the hypothesis, the k threshold,
  • 40:34the toxicity and efficacy scenarios scenario,
  • 40:38we see that dose three tends to be favored
  • 40:43in terms of allocation, where the highest media allocation,
  • 40:4826.7 and going all the way to 33.3 for the 75th percentile.
  • 40:57In terms of efficacy estimation, dose number three
  • 41:04or if you remember when we specify the true mean
  • 41:07efficacy was 40, the median estimated efficacy
  • 41:12in this case is 39.75,
  • 41:15the 75 percentile goes all the way to 45,
  • 41:19but of course that will be improved with,
  • 41:23as the sample size increases.
  • 41:28So in conclusion, what does iAdapt proposals?
  • 41:37It's an option, it's a viable option
  • 41:40for incorporating toxicity and efficacy outcomes,
  • 41:44especially for immunotherapy trials.
  • 41:49The novelty is in the designing allows
  • 41:56to model toxicity, both of binary
  • 42:01and also as quasi-continuous measures
  • 42:04and this was actually updated this year in the package
  • 42:08to use the several types of toxicities and several grades,
  • 42:17that continuous efficacy outcome
  • 42:22is very relevant for immunotherapies and I really showed
  • 42:26the example from the trial with T-cell persistence,
  • 42:29it is a relevant biomarker, but you can use for example,
  • 42:35absolute counts, you can use a full changes,
  • 42:39so design is flexible in incorporating
  • 42:41other continuous outcomes.
  • 42:43As far as I know, this is the only design at this point
  • 42:48that uses continuous efficacy outcomes.
  • 42:53So in terms of operating characteristics,
  • 42:58the design as well and allocating,
  • 43:00skewing the allocation to optimal doses,
  • 43:04estimation is marginally improved depends of course,
  • 43:08on the level of various and the sample size
  • 43:11and if anybody wants to try, you can use the R package,
  • 43:14you can use the Shiny app to simulate
  • 43:18to look at the behavior of different scenarios
  • 43:22to put that in trial and of course,
  • 43:26to use it to run the trial.
  • 43:30I'd like to thank two former students, Alyssa and Laura,
  • 43:34that helped in uploading the R package
  • 43:40and Laura has created the Shiny app.
  • 43:43And I do have some references in case you're interested,
  • 43:47but I can also share the slides later on.
  • 43:53So I think that is it and I wanted to allow some time
  • 43:57for questions and comments and feedback from you.
  • 44:02Thank you so much.
  • 44:06<v ->Thank you very much professor.</v>
  • 44:07(applauds)
  • 44:13Do you ave any questions in the room here?
  • 44:28Does anyone from Zoom have any questions?
  • 44:35<v ->Cody, thank you for the presentation,</v>
  • 44:37I think it's very useful to talk her way,
  • 44:40as well as my future like your possible designs
  • 44:46of the trials related to immunotherapy, I have one question.
  • 44:53So you mentioned toxicity and...
  • 45:01Does anywhere in the design actually dependent
  • 45:08on the independence of toxicity and efficacy profile?
  • 45:17<v ->It's a great point to talking</v>
  • 45:18if we actually modeled jointly toxicity and efficacy.
  • 45:23<v ->So I'm actually looking at the slide</v>
  • 45:26that you have the toxicity and like also efficacy profile,
  • 45:33I think like say if we have an ordinal categorical,
  • 45:37lets say for example, like say we pick a number three
  • 45:42because it's intolerable toxicity
  • 45:47and maximize the efficacy, right?
  • 45:50And anything above that will be too much
  • 45:52and anything below that will be like,
  • 45:55say not like effective enough.
  • 46:00So if they are actually,
  • 46:04how about certain joint distribution,
  • 46:09is there any thing we can do in order to like,
  • 46:13'cause that actually affect the simulation much?
  • 46:18<v ->So the current model does not account</v>
  • 46:22for the joint distribution,
  • 46:24it models toxicity and efficacy separately.
  • 46:27But as a next step we can look under to try to model
  • 46:35that dependency between toxicity and efficacy,
  • 46:38and especially for this novel agents,
  • 46:41we've seen that most of the times toxicity as is related
  • 46:48to efficacy, a stronger ethical, a stronger response
  • 46:54does come with some higher levels of toxicity,
  • 47:00but the current model does not look,
  • 47:02they twist them independent.
  • 47:06<v ->So do we, so is there, do you consider any,</v>
  • 47:11like penalty for example,
  • 47:12like say when there isn't such a good, like a compromise
  • 47:16between like minimizing toxicity
  • 47:20while maximizing efficacy, right?
  • 47:23So in the demonstration we have a compromise,
  • 47:26which is dose level number three,
  • 47:30if there's, for example, if there is like a conflict
  • 47:34between dose two and we can, like we don't really have,
  • 47:39like the obvious optimal, like say optimized solution.
  • 47:46Do we constantly there, like, for example, penalties
  • 47:49or do we always pay for like toxicity,
  • 47:53like say minimizing toxicity over like,
  • 47:57say maximizing efficacy?
  • 48:01<v ->And then think it's always, I think so for example,</v>
  • 48:07in that situation, so you could actually take both
  • 48:10dose three and dose number four,
  • 48:14you could consider both to be considered for future trials.
  • 48:20So, it's not the definite that the dose selected,
  • 48:30that you're always gonna reach a minimum of toxicity
  • 48:33and maximum efficacy, but you can look at different options
  • 48:39with as long as toxicity is acceptable,
  • 48:44you can consider maybe in phase-two
  • 48:47to look at randomized trial, look at dose level combo one
  • 48:52and dose levels combo-two based on efficacy
  • 48:55and we've actually seen this in a lot of trials,
  • 49:00the immune check point inhibitors review that I talked,
  • 49:05that it's now in progress, we looked at phase-one and two
  • 49:09and the rate of success and the design that are being used
  • 49:12and the doses that are being carried forward from phase-one
  • 49:16and phase-two, and surprisingly only 30%,
  • 49:21in 30% of phase-two trials the MTD was used from phase-one,
  • 49:30the rest either they use a lower dose
  • 49:33or they use a higher dose, but not the MTD.
  • 49:38So absolutely we can have like a range,
  • 49:42because if you think about it,
  • 49:43we have a limited sample size, right?
  • 49:46We need more information for efficacy,
  • 49:48so to complete the clear, the winner based on efficacy
  • 49:53might not be sufficient at this level.
  • 49:57<v ->Okay, great, thank you.</v>
  • 49:58So the answer is before phase-three
  • 50:01and as long as it's below the MTD,
  • 50:04the efficacy is important, is more important to prove,
  • 50:08like, say to move on to next stage.
  • 50:10Thank you.
  • 50:11<v Codruta>Yes.</v>
  • 50:29Just connection.
  • 50:30(chuckles)
  • 50:34<v Moderator>Sorry, we're still having a little weird audio</v>
  • 50:37issues obviously,
  • 50:45but does anybody in the room have any other questions
  • 50:48for the professor?
  • 51:02Or even we end the Zoom.
  • 51:11<v Wei>Hello, we have a question there.</v>
  • 51:16<v Moderator>Hold on.</v>
  • 51:21<v Student>Hi professor, I know we probably mentioned</v>
  • 51:22this already, but I probably didn't typed that,
  • 51:26can you repeat, maybe repeat what it was,
  • 51:28what do you consider would be like an advantage
  • 51:32of having a continuous efficacy
  • 51:35compares non-continuous efficacy in your model?
  • 51:40<v ->Yes, lots of information, so a lot of the lines</v>
  • 51:48are looking at the efficacy as a binary or ordinal,
  • 51:53there is actually one,
  • 51:55I don't know if you've heard of the Boyne,
  • 51:57that's also was published for immunotherapies
  • 52:00and that's using you take the efficacy levels
  • 52:06and you either dichotomized to represent
  • 52:09what is a successful or promising efficacy versus not,
  • 52:13and you pretty much modeled the probability of a response,
  • 52:18right one versus zero or at an ordinal level.
  • 52:22Number one, I think we were losing some information
  • 52:25when we do this categorization,
  • 52:28number two might be difficult to actually establish
  • 52:32this cutoffs and what represents a success
  • 52:35or how do we partition this efficacy range
  • 52:38for this novel agents.
  • 52:40So by looking at the continuous values,
  • 52:43we make the most out that information and we let it on,
  • 52:48we modeled it as such,
  • 52:54plus in the last couple of years,
  • 52:58this T-cell persistence has been shown
  • 53:01to be a promising biomarker.
  • 53:03So it's right on par with our proposal.
  • 53:18I know this might be a tough topic to digest for students
  • 53:24with early finding it's not such a...
  • 53:29It's a (chuckles) framework on its own.
  • 53:33So maybe not that everybody's familiar
  • 53:38with the whole terminology on the landscape.
  • 53:48<v Student>During that stimulation, you specifically...</v>
  • 53:52So the toxicity was stimulated
  • 53:55from a continuity distributions,
  • 53:58is there any specific reason
  • 53:59why you choose these distribution versus there's,
  • 54:03and if we similarly from a different distribution,
  • 54:08well, how about different conclusion like,
  • 54:11well, there would be any dependence
  • 54:13between efficacy and toxicity.
  • 54:17<v ->Yes, so that's, and so in this case,</v>
  • 54:21the results that I showed you were for toxicity,
  • 54:25for binary toxicity, yes or no.
  • 54:27So in a cohort of three patients for each patient,
  • 54:31you observed either a zero or a one response,
  • 54:36given the binary structure, it makes sense to use
  • 54:39this Bernoulli right distribution
  • 54:42and that sums up to binomial zero or one.
  • 54:46In terms of dependency is with what Dr. Cheng
  • 54:50was mentioning, we did not specify any correlation
  • 54:55between toxicity and efficacy
  • 54:57and did not look at the joint distribution between the two,
  • 55:00we modeled them separate and probably
  • 55:09that would be a good point moving forward.
  • 55:12What's difficult is how do we, what would be interesting
  • 55:17is looking at different levels of correlation
  • 55:21and see how in this joint distribution,
  • 55:25how the results with change, if we would capture that.
  • 55:38<v Wei>Okay, so any more questions?</v>
  • 55:50Okay, so thank you Dr. Chuizan,
  • 55:53for your wonderful presentation.
  • 55:58<v ->Thank you.</v>
  • 55:59<v ->Thank you, and if you have any questions,</v>
  • 56:02please email me anytime.
  • 56:03(chuckles)
  • 56:04And I'm sorry that you (indistinct).
  • 56:10Okay, I'll see you shortly, bye.
  • 56:13<v Wei>Thank you.</v>