1
|
Paul S, Zhou S. Six events that shaped antibody approvals in oncology. Front Immunol 2025; 16:1533796. [PMID: 39995677 PMCID: PMC11847691 DOI: 10.3389/fimmu.2025.1533796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
A little over twenty-five years ago, the European Medicines Agency (EMA) and the Food and Drug Administration (FDA) approved the chimeric antibody rituximab which fundamentally altered the landscape of anti-cancer drugs. While only a few antibodies were approved in the immediate years that followed the rituximab approval, the last decade saw a wave of antibody-drug approvals in the oncology arena. In the last three years, the EMA and FDA greenlighted eighteen antibodies, the majority of them designed in the formats of antibody-drug conjugates (ADC) and bispecific antibodies (BsAb). While the use of ADC and BsAb formats and the current rapid pace of approvals appear routine and almost inevitable, such progress was thought to be quite improbable in the early days of therapeutic antibody development. To understand how we arrived at the current state of antibody development in oncology, we focus on six monumental events that shaped antibody approvals over the last two and half decades. We examine the circumstances that led to the approval of rituximab and trastuzumab, the first successful antibodies for the treatment of hematologic and solid cancers. We detail the generation of the ADC and BsAb formats that dramatically augmented antibody-mediated precision cytotoxicity. Finally, we explore the development of ipilimumab, the first immune checkpoint-inhibiting antibody that activates the immune system to kill cancer cells, and the discovery that allowed the use of checkpoint inhibitors across all cancer types based on the presence of genetic markers. Revisiting these key events provides critical insights into the process of antibody development in oncology.
Collapse
Affiliation(s)
- Suman Paul
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Ludwig Center for Cancer Genetics and Therapeutics, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shibin Zhou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Ludwig Center for Cancer Genetics and Therapeutics, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
2
|
Silva RB, Cheng B, Carvajal RD, Lee SM. Dose Individualization for Phase I Cancer Trials With Broadened Eligibility. Stat Med 2024; 43:5534-5547. [PMID: 39479896 DOI: 10.1002/sim.10264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024]
Abstract
Broadening eligibility criteria in cancer trials has been advocated to represent the intended patient population more accurately. The advantages are clear in terms of generalizability and recruitment, however there are some important considerations in terms of design for efficiency and patient safety. While toxicity may be expected to be homogeneous across these subpopulations, designs should be able to recommend safe and precise doses if subpopulations with different toxicity profiles exist. Dose-finding designs accounting for patient heterogeneity have been proposed, but existing methods assume that the source of heterogeneity is known. We propose a broadened eligibility dose-finding design to address the situation of unknown patient heterogeneity in phase I cancer clinical trials where eligibility is expanded, and multiple eligibility criteria could potentially lead to different optimal doses for patient subgroups. The design offers a two-in-one approach to dose-finding by simultaneously selecting patient criteria that differentiate the maximum tolerated dose (MTD), using stochastic search variable selection, and recommending the subpopulation-specific MTD if needed. Our simulation study compares the proposed design to the naive approach of assuming patient homogeneity and demonstrates favorable operating characteristics across a wide range of scenarios, allocating patients more often to their true MTD during the trial, recommending more than one MTD when needed, and identifying criteria that differentiate the patient population. The proposed design highlights the advantages of adding more variability at an early stage and demonstrates how assuming patient homogeneity can lead to unsafe or sub-therapeutic dose recommendations.
Collapse
Affiliation(s)
- Rebecca B Silva
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York
| | - Bin Cheng
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York
| | - Richard D Carvajal
- Medical Oncology, Northwell Health Cancer Institute, New Hyde Park, New York
| | - Shing M Lee
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York
| |
Collapse
|
3
|
Knopick P, Terman D, Riha N, Alvine T, Larson R, Badiou C, Lina G, Ballantyne J, Bradley D. Endogenous HLA-DQ8αβ programs superantigens (SEG/SEI) to silence toxicity and unleash a tumoricidal network with long-term melanoma survival. J Immunother Cancer 2021; 8:jitc-2020-001493. [PMID: 33109631 PMCID: PMC7592263 DOI: 10.1136/jitc-2020-001493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/17/2022] Open
Abstract
Background As the most powerful T cell agonists known, superantigens (SAgs) have enormous potential for cancer immunotherapy. Their development has languished due to high incidence (60%–80%) of seroreactive neutralizing antibodies in humans and tumor necrosis factor-α (TNFα)-mediated cardiopulmonary toxicity. Such toxicity has narrowed their therapeutic index while neutralizing antibodies have nullified their therapeutic effects. Methods Female HLA-DQ8 (DQA*0301/DQB*0302) tg mice expressing the human major histocompatibility complex II (MHCII) HLA-DQ8 allele on a high proportion of PBL, spleen and lymph node cells were used. In the established tumor model, staphylococcal enterotoxin G and staphylococcal enterotoxin I (SEG/ SEI) (50 µg each) were injected on days 6 and 9 following tumor inoculation. Lymphoid, myeloid cells and tumor cell digests from tumor tissue were assayed using flow cytometry or quantitated using a cytometric bead array. Tumor density, necrotic and viable areas were quantitated using the ImageJ software. Results In a discovery-driven effort to address these problems we introduce a heretofore unrecognized binary complex comprizing SEG/SEI SAgs linked to the endogenous human MHCII HLA-DQ8 allele in humanized mice. By contrast to staphylococcal enterotoxin A (SEA) and staphylococcal enterotoxin B (SEB) deployed previously in clinical trials, SEG and SEI does not exhibit neutralizing antibodies in humans or TNFα-mediated toxicity in humanized HLA-DQ8 mice. In the latter model wherein SAg behavior is known to be ‘human-like’, SEG/SEI induced a powerful tumoricidal response and long-term survival against established melanoma in 82% of mice. Other SAgs deployed in the same model displayed toxic shock. Initially, HLA-DQ8 mediated melanoma antigen priming, after which SEG/SEI unleashed a broad CD4+ and CD8+ antitumor network marked by expansion of melanoma reactive T cells and interferon-γ (IFNy) in the tumor microenvironment (TME). SEG/SEI further initiated chemotactic recruitment of tumor reactive T cells to the TME converting the tumor from ‘cold’ to a ‘hot’. Long-term survivors displayed remarkable resistance to parental tumor rechallenge along with the appearance of tumor specific memory and tumor reactive T memory cells. Conclusions Collectively, these findings show for the first time that the SEG/SEI-(HLA-DQ8) empowers priming, expansion and recruitment of a population of tumor reactive T cells culminating in tumor specific memory and long-term survival devoid of toxicity. These properties distinguish SEG/SEI from other SAgs used previously in human tumor immunotherapy. Consolidation of these principles within the SEG/SEI-(HLA-DQ8) complex constitutes a conceptually new therapeutic weapon with compelling translational potential.
Collapse
Affiliation(s)
- Peter Knopick
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - David Terman
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Nathan Riha
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Travis Alvine
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Riley Larson
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Cedric Badiou
- University of Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Gerard Lina
- University of Lyon 1 University Institute of Tecnology Lyon 1, Villeurbanne, Auvergne-Rhône-Alpes, France
| | | | - David Bradley
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| |
Collapse
|
4
|
Kurzrock R, Lin CC, Wu TC, Hobbs BP, Pestana RC, Hong DS. Moving Beyond 3+3: The Future of Clinical Trial Design. Am Soc Clin Oncol Educ Book 2021; 41:e133-e144. [PMID: 34061563 DOI: 10.1200/edbk_319783] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Misgivings have been raised about the operating characteristics of the canonical 3+3 dose-escalation phase I clinical trial design. Yet, the traditional 3+3 design is still the most commonly used. Although it has been implied that adhering to this design is due to a stubborn reluctance to adopt change despite other designs performing better in hypothetical computer-generated simulation models, the continued adherence to 3+3 dose-escalation phase I strategies is more likely because these designs perform the best in the real world, pinpointing the correct dose and important side effects with an acceptable degree of precision. Beyond statistical simulations, there are little data to refute the supposed shortcomings ascribed to the 3+3 method. Even so, to address the unique nuances of gene- and immune-targeted compounds, a variety of inventive phase 1 trial designs have been suggested. Strategies for developing these therapies have launched first-in-human studies devised to acquire a breadth of patient data that far exceed the size of a typical phase I design and blur the distinction between dose selection and efficacy evaluation. Recent phase I trials of promising cancer therapies assessed objective tumor response and durability at various doses and schedules as well as incorporated multiple expansion cohorts spanning a variety of histology or biomarker-defined tumor subtypes, sometimes resulting in U.S. Food and Drug Administration approval after phase I. This article reviews recent innovations in phase I design from the perspective of multiple stakeholders and provides recommendations for future trials.
Collapse
Affiliation(s)
- Razelle Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsung-Che Wu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Brian P Hobbs
- Department of Population Health, Dell Medical School, University of Texas at Austin, Austin, TX
| | - Roberto Carmagnani Pestana
- Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
5
|
Soeny K, Bogacka B, Jones B. Model based dose personalization in clinical trials. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 201:105957. [PMID: 33588339 DOI: 10.1016/j.cmpb.2021.105957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/26/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND AND OBJECTIVE Personalized medicine is an important area of medical research which consists of designing therapies specifically for a patient or a group of patients. For drugs having a narrow therapeutic index or for vulnerable patients, methods such as therapeutic drug monitoring are used in a hospital setting to ensure that the blood concentration of the drug is maintained within a pre-decided range. However, such methods can not be used for drugs which are still in the developmental phase since, generally, insufficient information is available about the pharmacokinetic behaviour of the drug. METHODS In this paper, we present a new methodology for explicit optimization of dose regimens during the course of the pharmacokinetic studies such that the resultant blood concentration of the drug in each subject is maintained around a desired target concentration or within a target range. RESULTS We demonstrate that our algorithm is able to achieve the clinical objective of PK estimation while simultaneously individualizing the dose to every subject in the trial. Our algorithm computes dose regimens that, on average, have a relative efficiency of 97% with a standard deviation of less than 5%. The results show that the algorithm can be relied upon to ensure that the subjects in the trial are minimally over- and under-exposed to the test therapy. CONCLUSIONS The proposed methodology can assist in ensuring correct dosing to each subject in a clinical trial so that each subject receives only the intended exposure to the drug while simultaneously estimating the PK profile of the drug. Our methodology can also be applied in randomized concentration-controlled trials where maintenance of the target concentration in the subjects is a fundamental requirement for conducting these trials.
Collapse
Affiliation(s)
- Kabir Soeny
- School of Mathematical Sciences, Queen Mary University of London, UK.
| | - Barbara Bogacka
- School of Mathematical Sciences, Queen Mary University of London, UK
| | | |
Collapse
|
6
|
Zhang W, Wang X, Muthukumarana S, Yang P. A continual reassessment method without undue risk of toxicity. COMMUN STAT-SIMUL C 2021. [DOI: 10.1080/03610918.2021.1877306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Weijia Zhang
- Chongqing Key Laboratory of Social Economy and Applied Statistics, College of Mathematics and Statistics, Chongqing Technology and Business University, Chongqing, P. R. China
| | - Xikui Wang
- Warren Centre for Actuarial Studies and Research, I.H. Asper School of Business, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Saman Muthukumarana
- Department of Statistics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Po Yang
- Department of Statistics, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
7
|
Burnett T, Mozgunov P, Pallmann P, Villar SS, Wheeler GM, Jaki T. Adding flexibility to clinical trial designs: an example-based guide to the practical use of adaptive designs. BMC Med 2020; 18:352. [PMID: 33208155 PMCID: PMC7677786 DOI: 10.1186/s12916-020-01808-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Adaptive designs for clinical trials permit alterations to a study in response to accumulating data in order to make trials more flexible, ethical, and efficient. These benefits are achieved while preserving the integrity and validity of the trial, through the pre-specification and proper adjustment for the possible alterations during the course of the trial. Despite much research in the statistical literature highlighting the potential advantages of adaptive designs over traditional fixed designs, the uptake of such methods in clinical research has been slow. One major reason for this is that different adaptations to trial designs, as well as their advantages and limitations, remain unfamiliar to large parts of the clinical community. The aim of this paper is to clarify where adaptive designs can be used to address specific questions of scientific interest; we introduce the main features of adaptive designs and commonly used terminology, highlighting their utility and pitfalls, and illustrate their use through case studies of adaptive trials ranging from early-phase dose escalation to confirmatory phase III studies.
Collapse
Affiliation(s)
- Thomas Burnett
- Department of Mathematics and Statistics, Lancaster University, Fylde College, Lancaster, LA1 4YF UK
| | - Pavel Mozgunov
- Department of Mathematics and Statistics, Lancaster University, Fylde College, Lancaster, LA1 4YF UK
| | - Philip Pallmann
- Centre for Trials Research, College of Biomedical & Life Sciences, Cardiff University, Cardiff, UK
| | - Sofia S. Villar
- MRC Biostatistics Unit, University of Cambridge School of Clinical Medicine, Cambridge Institute of Public Health, Forvie Site, Robinson Way, Cambridge Biomedical Campus, Cambridge, CB2 0SR UK
| | - Graham M. Wheeler
- Cancer Research UK & UCL Cancer Trials Centre, University College London, 90 Tottenham Court Road, London, W1T 4TJ UK
| | - Thomas Jaki
- Department of Mathematics and Statistics, Lancaster University, Fylde College, Lancaster, LA1 4YF UK
- MRC Biostatistics Unit, University of Cambridge School of Clinical Medicine, Cambridge Institute of Public Health, Forvie Site, Robinson Way, Cambridge Biomedical Campus, Cambridge, CB2 0SR UK
| |
Collapse
|
8
|
Domenicano I, Ventz S, Cellamare M, Mak RH, Trippa L. Bayesian uncertainty‐directed dose finding designs. J R Stat Soc Ser C Appl Stat 2019. [DOI: 10.1111/rssc.12355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- I. Domenicano
- University of Rome “La Sapienza” Italy
- Dana–Farber Cancer Institute Boston USA
| | - S. Ventz
- Dana–Farber Cancer Institute Boston
- Harvard School of Public Health Boston USA
| | - M. Cellamare
- Dana–Farber Cancer Institute Boston
- Harvard School of Public Health Boston USA
| | - R. H. Mak
- Dana–Farber Cancer Institute Boston
- Harvard Medical School Boston USA
| | - L. Trippa
- Dana–Farber Cancer Institute Boston
- Harvard School of Public Health Boston USA
| |
Collapse
|
9
|
Diniz MA, Tighiouart M, Rogatko A. Comparison between continuous and discrete doses for model based designs in cancer dose finding. PLoS One 2019; 14:e0210139. [PMID: 30625194 PMCID: PMC6326565 DOI: 10.1371/journal.pone.0210139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 12/18/2018] [Indexed: 11/18/2022] Open
Abstract
Despite of an extensive statistical literature showing that discretizing continuous variables results in substantial loss of information, categorization of continuous variables has been a common practice in clinical research and in cancer dose finding (phase I) clinical trials. The objective of this study is to quantify the loss of information incurred by using a discrete set of doses to estimate the maximum tolerated dose (MTD) in phase I trials, instead of a continuous dose support. Escalation With Overdose Control and Continuous Reassessment Method were used because they are model-based designs where dose can be specified either as continuous or as a set of discrete levels. Five equally spaced sets of doses with different interval lengths and three sample sizes with sixteen scenarios were evaluated to compare the operating characteristics between continuous and discrete dose designs by Monte Carlo simulation. Loss of information was quantified by safety and efficiency measures. We conclude that if there is insufficient knowledge about the true MTD value, as commonly happens in phase I clinical trials, a continuous dose scheme minimizes information loss. If one is required to implement a design using discrete doses, then a scheme with 9 to 11 doses may yield similar results to the continuous dose scheme.
Collapse
Affiliation(s)
- Márcio Augusto Diniz
- Biostatistics and Bionformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California, United States of America
- * E-mail:
| | - Mourad Tighiouart
- Biostatistics and Bionformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California, United States of America
| | - André Rogatko
- Biostatistics and Bionformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California, United States of America
| |
Collapse
|
10
|
Tighiouart M. Two-stage design for phase I-II cancer clinical trials using continuous dose combinations of cytotoxic agents. J R Stat Soc Ser C Appl Stat 2019; 68:235-250. [PMID: 30745708 PMCID: PMC6368405 DOI: 10.1111/rssc.12294] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We present a two-stage phase I/II design of a combination of two drugs in cancer clinical trials. The goal is to estimate safe dose combination regions with a desired level of efficacy. In stage I, conditional escalation with overdose control is used to allocate dose combinations to successive cohorts of patients and the maximum tolerated dose curve is estimated as a function of Bayes estimates of the model parameters. In stage II, we propose a Bayesian adaptive design for conducting the phase II trial to determine dose combination regions along the MTD curve with a desired level of efficacy. The methodology is evaluated by extensive simulations and application to a real trial.
Collapse
|
11
|
Moni MM, Alam MI. Patient-specific dose finding in phase I clinical trials. J Appl Stat 2018. [DOI: 10.1080/02664763.2018.1428735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Md. Moniruzzaman Moni
- Institute of Statistical Research and Training, University of Dhaka, Dhaka, Bangladesh
| | - M. Iftakhar Alam
- Institute of Statistical Research and Training, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
12
|
Cotterill A, Jaki T. Dose-escalation strategies which use subgroup information. Pharm Stat 2018; 17:414-436. [PMID: 29900666 PMCID: PMC6175353 DOI: 10.1002/pst.1860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 01/30/2018] [Accepted: 02/26/2018] [Indexed: 12/04/2022]
Abstract
Dose-escalation trials commonly assume a homogeneous trial population to identify a single recommended dose of the experimental treatment for use in future trials. Wrongly assuming a homogeneous population can lead to a diluted treatment effect. Equally, exclusion of a subgroup that could in fact benefit from the treatment can cause a beneficial treatment effect to be missed. Accounting for a potential subgroup effect (ie, difference in reaction to the treatment between subgroups) in dose-escalation can increase the chance of finding the treatment to be efficacious in a larger patient population. A standard Bayesian model-based method of dose-escalation is extended to account for a subgroup effect by including covariates for subgroup membership in the dose-toxicity model. A stratified design performs well but uses available data inefficiently and makes no inferences concerning presence of a subgroup effect. A hypothesis test could potentially rectify this problem but the small sample sizes result in a low-powered test. As an alternative, the use of spike and slab priors for variable selection is proposed. This method continually assesses the presence of a subgroup effect, enabling efficient use of the available trial data throughout escalation and in identifying the recommended dose(s). A simulation study, based on real trial data, was conducted and this design was found to be both promising and feasible.
Collapse
Affiliation(s)
- Amy Cotterill
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| | - Thomas Jaki
- Medical and Pharmaceutical Statistics Research Unit, Department of Mathematics and StatisticsLancaster UniversityLancasterUK
| |
Collapse
|
13
|
Wheeler GM, Sweeting MJ, Mander AP. Toxicity-dependent feasibility bounds for the escalation with overdose control approach in phase I cancer trials. Stat Med 2017; 36:2499-2513. [PMID: 28295513 PMCID: PMC5462100 DOI: 10.1002/sim.7280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 01/19/2017] [Accepted: 02/18/2017] [Indexed: 11/09/2022]
Abstract
Phase I trials of anti-cancer therapies aim to identify a maximum tolerated dose (MTD), defined as the dose that causes unacceptable toxicity in a target proportion of patients. Both rule-based and model-based methods have been proposed for MTD recommendation. The escalation with overdose control (EWOC) approach is a model-based design where the dose assigned to the next patient is one that, given all available data, has a posterior probability of exceeding the MTD equal to a pre-specified value known as the feasibility bound. The aim is to conservatively dose-escalate and approach the MTD, avoiding severe overdosing early on in a trial. The EWOC approach has been applied in practice with the feasibility bound either fixed or varying throughout a trial, yet some of the methods may recommend incoherent dose-escalation, that is, an increase in dose after observing severe toxicity at the current dose. We present examples where varying feasibility bounds have been used in practice, and propose a toxicity-dependent feasibility bound approach that guarantees coherent dose-escalation and incorporates the desirable features of other EWOC approaches. We show via detailed simulation studies that the toxicity-dependent feasibility bound approach provides improved MTD recommendation properties to the original EWOC approach for both discrete and continuous doses across most dose-toxicity scenarios, with comparable performance to other approaches without recommending incoherent dose escalation. © 2017 The Authors. Statistics in Medicine Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- Graham M. Wheeler
- Cancer Research UK and UCL Cancer Trials CentreUniversity College LondonU.K.
- MRC Biostatistics Unit Hub for Trials Methodology ResearchCambridge Institute of Public HealthCambridgeU.K.
| | - Michael J. Sweeting
- Cardiovascular Epidemiology UnitStrangeways Research Laboratory University of CambridgeU.K.
| | - Adrian P. Mander
- MRC Biostatistics Unit Hub for Trials Methodology ResearchCambridge Institute of Public HealthCambridgeU.K.
| |
Collapse
|
14
|
Si SY, Liu JL, Liu JL, Xu BX, Li JZ, Qin YY, Song SJ. Recombinant adenovirus of SEA and CD80 genes driven by MMRE and mouse TERT promoter induce effective antitumor immune responses against different types of tumor cells in vitro and in vivo. Oncol Rep 2017; 37:3037-3045. [PMID: 28393253 DOI: 10.3892/or.2017.5563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/06/2017] [Indexed: 11/05/2022] Open
Abstract
Staphylococcus enterotoxin A (SEA) is a powerful immunostimulant and can stimulate T cells bearing certain T-cell receptor β-chain variable regions when bound to major histocompatibility complex II molecules. SEA is widely used in research of antitumor therapy. The low affinity T-cell receptor (TCR) interaction with SEA in the absence of MHC class II antigens is sufficient for the induction of cytotoxicity but requires additional CD28/B7 signaling to result in proliferation of resting T cells. In this study, we constructed recombinant adenovirus (named as Ad-MMRE-mTERT-BIS) carrying membrane-expressing SEA (named as SEAtm) and CD80 driven by Myc-Max response elements (MMRE) and mouse telomerase reverse transcriptase (mTERT) promoter to reduce toxicity and to improve safety and efficiency. We demonstrated that Ad-MMRE-mTERT-BIS could make SEAtm and CD80 to co-express highly on the surface of Hepa1-6 and B16 cells, at low level on the surface of CT26 cells, but not in NIH3T3. Hepa1-6 and B16 cells infected by the recombinant adenovirus induced proliferation of CD4+ and CD8+ T cells and increased cytokine [interleukin (IL)-2, tumor necrosis factor (TNF)-α, interferon (IFN)-γ] production in vitro. Intratumoral injection of Ad-MMRE-mTERT-BIS in hepatoma and melanoma mouse models induced tumor-specific cytotoxic T cells in the spleen. Moreover, hepatoma and melanoma xenografts were suppressed by treatment with Ad-MMRE-mTERT-BIS and the survival time of treated mice was prolonged. These findings suggest that recombinant adenovirus of SEA and CD80 genes driven by mTERT promoter could induce effective antitumor immune responses against different kinds of tumor cells in vitro and in vivo.
Collapse
Affiliation(s)
- Shao-Yan Si
- Center for Special Medicine and Experimental Research, 306th Hospital of PLA, Beijing 100101, P.R. China
| | - Jun-Li Liu
- Center for Special Medicine and Experimental Research, 306th Hospital of PLA, Beijing 100101, P.R. China
| | - Jun-Lian Liu
- Department of Dermatology, 306th Hospital of PLA, Beijing 100101, P.R. China
| | - Bing-Xin Xu
- Center for Special Medicine and Experimental Research, 306th Hospital of PLA, Beijing 100101, P.R. China
| | - Jian-Zhong Li
- Center for Special Medicine and Experimental Research, 306th Hospital of PLA, Beijing 100101, P.R. China
| | - Ya-Ya Qin
- Center for Special Medicine and Experimental Research, 306th Hospital of PLA, Beijing 100101, P.R. China
| | - Shu-Jun Song
- Center for Special Medicine and Experimental Research, 306th Hospital of PLA, Beijing 100101, P.R. China
| |
Collapse
|
15
|
Stern PL, Harrop R. 5T4 oncofoetal antigen: an attractive target for immune intervention in cancer. Cancer Immunol Immunother 2017; 66:415-426. [PMID: 27757559 PMCID: PMC11029567 DOI: 10.1007/s00262-016-1917-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/12/2016] [Indexed: 01/18/2023]
Abstract
The natural history of a patient's cancer is often characterised by genetic diversity and sequential sweeps of clonal dominance. It is therefore not surprising that identifying the most appropriate tumour-associated antigen for targeted intervention is challenging. The 5T4 oncofoetal antigen was identified by searching for surface molecules shared between human trophoblast and cancer cells with the rationale that they may function to allow survival of the foetus as a semi-allograft in the mother or a tumour in its host. The 5T4 protein is expressed by many different cancers but rarely in normal adult tissues. 5T4 molecules are 72 kD, heavily N-glycosylated proteins with several leucine-rich repeats which are often associated with protein-protein interactions. 5T4 expression is associated with the directional movement of cells through epithelial mesenchymal transition, potentiation of CXCL12/CXCR4 chemotaxis and inhibition of canonical Wnt/beta-catenin while favouring non-canonical pathway signalling; all processes which help drive the spread of cancer cells. The selective pattern of 5T4 tumour expression, association with a tumour-initiating phenotype plus a mechanistic involvement with cancer spread have underwritten the clinical development of different immunotherapeutic strategies including a vaccine, a tumour-targeted superantigen and an antibody drug conjugate. In addition, a chimeric antigen receptor T cell approach targeting 5T4 expressing tumour cells is in pre-clinical development. A key challenge will include how best to combine each 5T4 targeted immunotherapy with the most appropriate standard of care treatment (or adjunct therapy) to maximise the recovery of immune control and ultimately eliminate the tumour.
Collapse
Affiliation(s)
- Peter L Stern
- Institute of Cancer Studies, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | - Richard Harrop
- Oxford BioMedica Plc, Windrush Court, Transport Way, Oxford, OX4 6LT, UK.
| |
Collapse
|
16
|
Agheli R, Emkanian B, Halabian R, Fallah Mehrabadi J, Imani Fooladi AA. Recombinant Staphylococcal Enterotoxin Type A Stimulate Antitumoral Cytokines. Technol Cancer Res Treat 2017; 16:125-132. [PMID: 27884943 PMCID: PMC5616123 DOI: 10.1177/1533034616679344] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/02/2016] [Accepted: 10/12/2016] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND About 20 different types of staphylococcal enterotoxins are produced by Staphylococcus aureus, in which type A is more common in food poisoning syndrome. Also staphylococcal enterotoxin A superantigen is a potent inducer of cytotoxic T lymphocyte activity and cytokine production and could stimulate T cells containing T-cell receptor beta chain domains when binding to major histocompatibility complex class II molecules. Hence, it is an important reagent in cancer immunotherapy. METHODS For the construction of pET-21a/ entA cassette, the staphylococcal enterotoxin type A gene was isolated from S aureus strain HN2, cloned into pET-21a, and introduced into Escherichia coli strain BL-21(DE3). Consequently, Western blot analysis showed pET-21a/ entA cassette expression inserted entA gene successfully. It is the first prompt using a pET-21a as a cloning vector for entA gene and expression of construct in BL-21(DE3). In addition, this study examined the ability of standard staphylococcal enterotoxin A and cloned staphylococcal enterotoxin A to activate T cells in vitro. Lymphocyte cells derived from lymph node BALB/c mice were exposed to standard staphylococcal enterotoxin A and cloned staphylococcal enterotoxin (1.10, 102,103, and 104 ng/µL) in order to evaluate the magnitude of proliferation, activation, and apoptosis of lymphocyte cells based on MTT and apoptosis assays, respectively. RESULTS Our investigation showed that the function of cloned staphylococcal enterotoxin A was same as standard staphylococcal enterotoxin A, and the optimal concentration for the activation of lymphocyte cells and induction of apoptosis was 100 ng/µL and 1000 ng/µL ( P < .05), respectively. Quantification of cytokines clearly showed that lymphocyte cells exposed to standard staphylococcal enterotoxin A and cloned staphylococcal enterotoxin A significantly secreted higher interferon γ and tumor necrosis factor α compared to control. CONCLUSION According to our results, the biological activity of standard staphylococcal enterotoxin A and cloned staphylococcal enterotoxin A is identical; therefore, these procedures may be approved as an efficient method to express and purify this protein in a large scale.
Collapse
Affiliation(s)
- Reza Agheli
- Applied Microbiology, Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Bijan Emkanian
- Applied Microbiology, Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology, Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jalil Fallah Mehrabadi
- Applied Microbiology, Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology, Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Abstract
Background In phase I cancer clinical trials, adjustment for patient differences in toxicity susceptibility can be carried out with stratification into risk groups. Separate trials conducted for each risk group can lead to conflicting decisions, in which higher doses are recommended for higher risk groups. Designs which covariate adjust often require assumptions that clinicians may be uncomfortable with. Methods We extend up-and-down designs, isotonic designs and the continual reassessment method (CRM) to multiple risk groups with two-way isotonic regression. The only assumption about the groups is that they can be ordered according to their toxicity risk. The first two extensions, in particular, are nonparametric and easy for clinicians to understand. Results Simulations were based on an ongoing helical tomotherapy trial. Seven different toxicity scenarios were considered. The proposed methods compared favorably to a covariate adjusted CRM. The extended up-and-down designs inherited the conservativeness from the original designs. Conclusion Our experience demonstrates that the escalation rules of multiple risk groups can be linked, without a parametric assumption about the group toxicity curve, to borrow strength and to ensure nonconflicting dosage recommendations.
Collapse
Affiliation(s)
- Zhilong Yuan
- Department of Statistics, University of Wisconsin, Madison, WI, USA.
| | | |
Collapse
|
18
|
Abstract
A desirable property of any dose-escalation strategy for phase I oncology trials is coherence: if the previous patient experienced a toxicity, a higher dose is not recommended for the next patient; similarly, if the previous patient did not experience a toxicity, a lower dose is not recommended for the next patient. The escalation with overdose control (EWOC) approach is a model-based design that has been applied in practice, under which the dose assigned to the next patient is the one that, given all available data, has a posterior probability of exceeding the maximum tolerated dose equal to a pre-specified value known as the feasibility bound. Several methodological and applied publications have considered the EWOC approach with both feasibility bounds fixed and increasing throughout the trial. Whilst the EWOC approach with fixed feasibility bound has been proven to be coherent, some proposed methods of increasing the feasibility bound regardless of toxicity outcomes of patients can lead to incoherent dose-escalation. This paper formalises a proof that incoherent dose-escalation can occur if the feasibility bound is increased without consideration of preceding toxicity outcomes, and shows via simulation studies that only small increases in the feasibility bound are required for incoherent dose-escalations to occur.
Collapse
|
19
|
Elkord E, Burt DJ, Sundstedt A, Nordle Ö, Hedlund G, Hawkins RE. Immunological response and overall survival in a subset of advanced renal cell carcinoma patients from a randomized phase 2/3 study of naptumomab estafenatox plus IFN-α versus IFN-α. Oncotarget 2015; 6:4428-39. [PMID: 25669986 PMCID: PMC4414201 DOI: 10.18632/oncotarget.2922] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/11/2014] [Indexed: 01/05/2023] Open
Abstract
Naptumomab estafenatox/ABR-217620/ANYARA (Nap) has been evaluated in clinical phase 1 and 2/3 studies. RCC patients in the phase 2/3 trial were randomized 1:1 in an open label study to receive Nap+IFN-α or IFN-α. In this study, we analyzed the UK patients for their immunological response in relation to prolonged overall survival (OS). We found that Nap-specific T cells were reduced after 3 treatment days in patients' peripheral blood. Levels of both Nap-specific CD4+ and CD8+ T cells were significantly higher 8 days after the first treatment. Patients with such pattern of reduction and expansion of Nap-binding T cells also showed increased levels of IL-2 and IFN-γ in plasma 3 hours after the first Nap treatment. In addition, Nap caused an increase of IL-6, IL-10 and TNF-α. The patients in the UK subset showed a tendency of OS benefit after Nap treatment. Most Nap treated patients with long OS had low baseline IL-6 and normal levels of anti-SEA/E-120 antibodies. Furthermore, patients with pronounced Nap induced IL-2 and T cell expansion had long OS. In conclusion, patients with low baseline IL-6 and normal anti-SEA/E-120 may respond well to Nap by T cell activation and expansion paving the way for anti-tumour effects.
Collapse
Affiliation(s)
- Eyad Elkord
- Department of Medical Oncology, Institute of Cancer Sciences, The University of Manchester, Manchester, UK.,Department of Medical Microbiology & Immunology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, UAE.,Biomedical Research Centre, School of Environment & Life Sciences, University of Salford, Salford, UK
| | - Deborah J Burt
- Department of Medical Oncology, Institute of Cancer Sciences, The University of Manchester, Manchester, UK
| | | | | | | | - Robert E Hawkins
- Department of Medical Oncology, Institute of Cancer Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
20
|
Rogatko A, Cook-Wiens G, Tighiouart M, Piantadosi S. Escalation with Overdose Control is More Efficient and Safer than Accelerated Titration for Dose Finding. ENTROPY 2015; 17:5288-5303. [PMID: 27156869 PMCID: PMC4859761 DOI: 10.3390/e17085288] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The standard 3 + 3 or "modified Fibonacci" up-and-down (MF-UD) method of dose escalation is by far the most used design in dose-finding cancer trials. However, MF-UD has always shown inferior performance when compared with its competitors regarding number of patients treated at optimal doses. A consequence of using less effective designs is that more patients are treated with doses outside the therapeutic window. In June 2012, the U S Food and Drug Administration (FDA) rejected the proposal to use Escalation with Overdose Control (EWOC), an established dose-finding method which has been extensively used in FDA-approved first in human trials and imposed a variation of the MF-UD, known as accelerated titration (AT) design. This event motivated us to perform an extensive simulation study comparing the operating characteristics of AT and EWOC. We show that the AT design has poor operating characteristics relative to three versions of EWOC under several practical scenarios. From the clinical investigator's perspective, lower bias and mean square error make EWOC designs preferable than AT designs without compromising safety. From a patient's perspective, uniformly higher proportion of patients receiving doses within an optimal range of the true MTD makes EWOC designs preferable than AT designs.
Collapse
Affiliation(s)
- André Rogatko
- Author to whom correspondence should be addressed; ; Tel.: +1-310-423-3316; Fax: +1-310-423-4020
| | | | | | | |
Collapse
|
21
|
Eisen T, Hedlund G, Forsberg G, Hawkins R. Naptumomab estafenatox: targeted immunotherapy with a novel immunotoxin. Curr Oncol Rep 2014; 16:370. [PMID: 24445502 PMCID: PMC3918406 DOI: 10.1007/s11912-013-0370-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Improvement of cancer therapy by introducing new concepts is still urgent even though there have been major advancements lately. Immunotherapy is well on the way to becoming an established tool in the cancer treatment armory. It seems that a combination of (1) activation of immune effector cells and selective targeting of them to tumors and (2) the inhibition of immune suppression often induced by the tumor itself are necessary to achieve the therapeutic goal. The immunotoxin naptumomab estafenatox was developed in an effort to activate and target the patient’s own T cells to their tumor, by fusing a superantigen (SAg) variant that activates T lymphocytes to the Fab moiety of a tumor-reactive monoclonal antibody. Naptumomab estafenatox targets the 5T4 tumor antigen, a 72-kDa oncofetal trophoblast protein expressed on many carcinomas, including renal cell carcinoma. The therapeutic effect is associated with activation of SAg-binding T cells. The SAg-binding T lymphocytes expand, differentiate to effector cells, and infiltrate the tumor. The therapeutic efficacy is most likely related to the dual mechanism of tumor cell killing: (1) direct lysis by cytotoxic T lymphocytes of tumor cells expressing the antigen recognized by the antibody moiety of the fusion protein and (2) secretion of cytokines eliminating antigen-negative tumor cell variants. Naptumomab estafenatox has been clinically tested in a range of solid tumors with focus on renal cell carcinoma. This review looks at the clinical experience with the new immunotoxin and its potential.
Collapse
Affiliation(s)
- Tim Eisen
- Cambridge University Health Partners, Addenbrooke's Hospital, Cambridge, UK,
| | | | | | | |
Collapse
|
22
|
Iasonos A, O'Quigley J. Adaptive dose-finding studies: a review of model-guided phase I clinical trials. J Clin Oncol 2014; 32:2505-11. [PMID: 24982451 DOI: 10.1200/jco.2013.54.6051] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE We provide a comprehensive review of adaptive phase I clinical trials in oncology that used a statistical model to guide dose escalation to identify the maximum-tolerated dose (MTD). We describe the clinical setting, practical implications, and safety of such applications, with the aim of understanding how these designs work in practice. METHODS We identified 53 phase I trials published between January 2003 and September 2013 that used the continual reassessment method (CRM), CRM using escalation with overdose control, or time-to-event CRM for late-onset toxicities. Study characteristics, design parameters, dose-limiting toxicity (DLT) definition, DLT rate, patient-dose allocation, overdose, underdose, sample size, and trial duration were abstracted from each study. In addition, we examined all studies in terms of safety, and we outlined the reasons why escalations occur and under what circumstances. RESULTS On average, trials accrued 25 to 35 patients over a 2-year period and tested five dose levels. The average DLT rate was 18%, which is lower than in previous reports, whereas all levels above the MTD had an average DLT rate of 36%. On average, 39% of patients were treated at the MTD, and 74% were treated at either the MTD or an adjacent level (one level above or below). CONCLUSION This review of completed phase I studies confirms the safety and generalizability of model-guided, adaptive dose-escalation designs, and it provides an approach for using, interpreting, and understanding such designs to guide dose escalation in phase I trials.
Collapse
Affiliation(s)
- Alexia Iasonos
- Alexia Iasonos, Memorial Sloan Kettering Cancer Center, New York, NY; and John O'Quigley, Université Paris VI, Paris, France.
| | - John O'Quigley
- Alexia Iasonos, Memorial Sloan Kettering Cancer Center, New York, NY; and John O'Quigley, Université Paris VI, Paris, France
| |
Collapse
|
23
|
Patterson KG, Dixon Pittaro JL, Bastedo PS, Hess DA, Haeryfar SMM, McCormick JK. Control of established colon cancer xenografts using a novel humanized single chain antibody-streptococcal superantigen fusion protein targeting the 5T4 oncofetal antigen. PLoS One 2014; 9:e95200. [PMID: 24736661 PMCID: PMC3988171 DOI: 10.1371/journal.pone.0095200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/25/2014] [Indexed: 01/21/2023] Open
Abstract
Superantigens (SAgs) are microbial toxins that cross-link T cell receptors with major histocompatibility class II (MHC-II) molecules leading to the activation of large numbers of T cells. Herein, we describe the development and preclinical testing of a novel tumor-targeted SAg (TTS) therapeutic built using the streptococcal pyrogenic exotoxin C (SpeC) SAg and targeting cancer cells expressing the 5T4 tumor-associated antigen (TAA). To inhibit potentially harmful widespread immune cell activation, a SpeC mutation within the high-affinity MHC-II binding interface was generated (SpeCD203A) that demonstrated a pronounced reduction in mitogenic activity, yet this mutant could still induce immune cell-mediated cancer cell death in vitro. To target 5T4+ cancer cells, we engineered a humanized single chain variable fragment (scFv) antibody to recognize 5T4 (scFv5T4). Specific targeting of scFv5T4 was verified. SpeCD203A fused to scFv5T4 maintained the ability to activate and induce immune cell-mediated cytotoxicity of colorectal cancer cells. Using a xenograft model of established human colon cancer, we demonstrated that the SpeC-based TTS was able to control the growth and spread of large tumors in vivo. This required both TAA targeting by scFv5T4 and functional SAg activity. These studies lay the foundation for the development of streptococcal SAgs as ‘next-generation’ TTSs for cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/immunology
- Antigens, Neoplasm/immunology
- Cell Line, Tumor
- Cell Proliferation
- Cell Transformation, Neoplastic
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Humans
- Immunotherapy/methods
- Mice
- Models, Molecular
- Protein Conformation
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Streptococcus/immunology
- Superantigens/genetics
- Superantigens/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Kelcey G. Patterson
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | | | - Peter S. Bastedo
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - David A. Hess
- Department of Physiology and Pharmacology, Western University, London Ontario, Canada
- Vascular Biology Research Group, Robarts Research Institute, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
24
|
Chen Z, Cui Y, Owonikoko TK, Wang Z, Li Z, Luo R, Kutner M, Khuri FR, Kowalski J. Escalation with overdose control using all toxicities and time to event toxicity data in cancer Phase I clinical trials. Contemp Clin Trials 2014; 37:322-32. [PMID: 24530487 DOI: 10.1016/j.cct.2014.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 01/27/2014] [Accepted: 02/05/2014] [Indexed: 11/18/2022]
Abstract
The primary purposes of Phase I cancer clinical trials are to determine the maximum tolerated dose (MTD) and the treatment schedule of a new drug. Phase I trials usually involve a small number of patients so that fully utilizing all toxicity information including time to event toxicity data is key to improving the trial efficiency and the accuracy of MTD estimation. Chen et al. proposed a novel normalized equivalent toxicity score (NETS) system to fully utilize multiple toxicities per patient instead of a binary indicator of dose limiting toxicity (DLT). Cheung and Chappell developed the time to toxicity event (TITE) approach to incorporate time to toxicity event data. Escalation with overdose control (EWOC) is an adaptive Bayesian Phase I design which can allow rapid dose escalation while controlling the probability of overdosing patients. In this manuscript, we use EWOC as a framework and integrate it with the NETS system and the TITE approach to develop an advanced Phase I design entitled EWOC-NETS-TITE. We have conducted simulation studies to compare its operating characteristics using selected derived versions of EWOC because EWOC itself has already been extensively compared with common Phase I designs [3]. Simulation results demonstrate that EWOC-NETS-TITE can substantially improve the trial efficiency and accuracy of MTD determination as well as allow patients to be entered in a staggered fashion to significantly shorten trial duration. Moreover, user-friendly software for EWOC-NETS-TITE is under development.
Collapse
Affiliation(s)
- Zhengjia Chen
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States; Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States.
| | - Ye Cui
- ICF International, 3 Corporate Square, NE, Suite 370, Atlanta, GA 30329, United States
| | - Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, United States
| | - Zhibo Wang
- Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States
| | - Zheng Li
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States
| | - Ruiyan Luo
- School of Public Health, Georgia State University, Atlanta, GA 30303, United States
| | - Michael Kutner
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, United States
| | - Jeanne Kowalski
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States; Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States
| |
Collapse
|
25
|
Sverdlov O, Wong WK, Ryeznik Y. Adaptive clinical trial designs for phase I cancer studies. STATISTICS SURVEYS 2014. [DOI: 10.1214/14-ss106] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
26
|
Hedlund G, Eriksson H, Sundstedt A, Forsberg G, Jakobsen BK, Pumphrey N, Rödström K, Lindkvist-Petersson K, Björk P. The tumor targeted superantigen ABR-217620 selectively engages TRBV7-9 and exploits TCR-pMHC affinity mimicry in mediating T cell cytotoxicity. PLoS One 2013; 8:e79082. [PMID: 24194959 PMCID: PMC3806850 DOI: 10.1371/journal.pone.0079082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 09/26/2013] [Indexed: 01/20/2023] Open
Abstract
The T lymphocytes are the most important effector cells in immunotherapy of cancer. The conceptual objective for developing the tumor targeted superantigen (TTS) ABR-217620 (naptumomab estafenatox, 5T4Fab-SEA/E-120), now in phase 3 studies for advanced renal cell cancer, was to selectively coat tumor cells with cytotoxic T lymphocytes (CTL) target structures functionally similar to natural CTL pMHC target molecules. Here we present data showing that the molecular basis for the anti-tumor activity by ABR-217620 resides in the distinct interaction between the T cell receptor β variable (TRBV) 7-9 and the engineered superantigen (Sag) SEA/E-120 in the fusion protein bound to the 5T4 antigen on tumor cells. Multimeric but not monomeric ABR-217620 selectively stains TRBV7-9 expressing T lymphocytes from human peripheral blood similar to antigen specific staining of T cells with pMHC tetramers. SEA/E-120 selectively activates TRBV7-9 expressing T lymphocytes resulting in expansion of the subset. ABR-217620 selectively triggers TRBV7-9 expressing cytotoxic T lymphocytes to kill 5T4 positive tumor cells. Furthermore, ABR-217620 activates TRBV7-9 expressing T cell line cells in the presence of cell- and bead-bound 5T4 tumor antigen. Surface plasmon resonance analysis revealed that ABR-217620 binds to 5T4 with high affinity, to TRBV7-9 with low affinity and to MHC class II with very low affinity. The T lymphocyte engagement by ABR-217620 is constituted by displaying high affinity binding to the tumor cells (KD approximately 1 nM) and with the mimicry of natural productive immune TCR-pMHC contact using affinities of around 1 µM. This difference in kinetics between the two components of the ABR-217620 fusion protein will bias the binding towards the 5T4 target antigen, efficiently activating T-cells via SEA/E-120 only when presented by the tumor cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Karin Rödström
- Experimental Medical Science, Lund University, Lund, Sweden
| | | | | |
Collapse
|
27
|
|
28
|
Chen Z, Wang Z, Wang H, Owonikoko TK, Kowalski J, Khuri FR. Interactive Software "Isotonic Design using Normalized Equivalent Toxicity Score (ID-NETS©TM)" for Cancer Phase I Clinical Trials. Open Med Inform J 2013; 7:8-17. [PMID: 23847695 PMCID: PMC3680993 DOI: 10.2174/1874431101307010008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 01/17/2013] [Accepted: 01/17/2013] [Indexed: 11/22/2022] Open
Abstract
Isotonic Design using Normalized Equivalent Toxicity Score (ID-NETS) is a novel Phase I design that integrates the novel toxicity scoring system originally proposed by Chen et al. [1] and the original Isotonic Design proposed by Leung et al. [2]. ID-NETS has substantially improved the accuracy of maximum tolerated dose (MTD) estimation and trial efficiency in the Phase I clinical trial setting by fully utilizing all toxicities experienced by each patient and treating toxicity response as a quasi-continuous variable instead of a binary indicator of dose limiting toxicity (DLT). To facilitate the incorporation of the ID-NETS method into the design and conduct of Phase I clinical trials, we have designed and developed a user-friendly software, ID-NETS(©TM), which has two functions: 1) Calculating the recommended dose for the subsequent patient cohort using available completed data; and 2) Performing simulations to obtain the operating characteristics of a trial designed with ID-NETS. Currently, ID-NETS(©TM)v1.0 is available for free download at http://winshipbbisr.emory.edu/IDNETS.html.
Collapse
Affiliation(s)
- Zhengjia Chen
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, USA ; Biostatistics and Bioinformatics Shared Resource at Winship Cancer Institute, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
29
|
Rogatko A, Ghosh P, Vidakovic B, Tighiouart M. Patient-Specific Dose Adjustment in the Cancer Clinical Trial Setting. Pharmaceut Med 2012. [DOI: 10.1007/bf03256730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
30
|
Dose escalation with overdose control using a quasi-continuous toxicity score in cancer Phase I clinical trials. Contemp Clin Trials 2012; 33:949-58. [PMID: 22561391 DOI: 10.1016/j.cct.2012.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 04/05/2012] [Accepted: 04/13/2012] [Indexed: 11/20/2022]
Abstract
Escalation with overdose control (EWOC) is a Bayesian adaptive design for selecting dose levels in cancer Phase I clinical trials while controlling the posterior probability of exceeding the maximum tolerated dose (MTD). EWOC has been used by clinicians to design many cancer Phase I clinical trials, see e.g. [1-4]. However, this design treats the toxicity response as a binary indicator of dose limiting toxicity (DLT) and does not account for the number and specific grades of toxicities experienced by patients during the trial. Chen et al. (2010) proposed a novel toxicity score system to fully utilize all toxicity information using a normalized equivalent toxicity score (NETS). In this paper, we propose to incorporate NETS into EWOC using a quasi-Bernoulli likelihood approach to design cancer Phase I clinical trials. We call the design escalation with overdose control using normalized equivalent toxicity score (EWOC-NETS). Simulation results show that this design has good operating characteristics and improves the accuracy of MTD, trial efficiency, therapeutic effect, and overdose control relative to EWOC which is used as a representative of designs treating toxicity response as a binary indicator of DLT. We illustrate the performance of this design using real trial data in identifying the Phase II dose.
Collapse
|
31
|
Escalation with Overdose Control Using Ordinal Toxicity Grades for Cancer Phase I Clinical Trials. JOURNAL OF PROBABILITY AND STATISTICS 2012. [DOI: 10.1155/2012/317634] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We extend a Bayesian adaptive phase I clinical trial design known as escalation with overdose control (EWOC) by introducing an intermediate grade 2 toxicity when assessing dose-limiting toxicity (DLT). Under the proportional odds model assumption of dose-toxicity relationship, we prove that in the absence of DLT, the dose allocated to the next patient given that the previously treated patient had a maximum of grade 2 toxicity is lower than the dose given to the next patient had the previously treated patient exhibited a grade 0 or 1 toxicity at the most. Further, we prove that the coherence properties of EWOC are preserved. Simulation results show that the safety of the trial is not compromised and the efficiency of the estimate of the maximum tolerated dose (MTD) is maintained relative to EWOC treating DLT as a binary outcome and that fewer patients are overdosed using this design when the true MTD is close to the minimum dose.
Collapse
|
32
|
Number of Patients per Cohort and Sample Size Considerations Using Dose Escalation with Overdose Control. JOURNAL OF PROBABILITY AND STATISTICS 2012. [DOI: 10.1155/2012/692725] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The main objective of cancer phase I clinical trials is to determine a maximum tolerated dose (MTD) of a new experimental treatment. In practice, most of these trials are designed so that three patients per cohort are treated at the same dose level. In this paper, we compare the safety and efficiency of trials using the escalation with overdose control (EWOC) scheme designed with three or only one patient per cohort. We show through simulations that the number of patients per cohort does not impact the proportion of patients given therapeutic doses, safety of the trial, and efficiency of the estimate of the MTD. Additionally, we present guidelines and tabulated values on the number of patients needed to design a phase I cancer clinical trial using EWOC to achieve a given accuracy of the estimate of the MTD.
Collapse
|
33
|
Kato M, Nakamura Y, Suda T, Ozawa Y, Inui N, Seo N, Nagata T, Koide Y, Kalinski P, Nakamura H, Chida K. Enhanced anti-tumor immunity by superantigen-pulsed dendritic cells. Cancer Immunol Immunother 2011; 60:1029-38. [PMID: 21519830 PMCID: PMC11029592 DOI: 10.1007/s00262-011-1015-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 03/31/2011] [Indexed: 12/25/2022]
Abstract
Staphylococcal enterotoxins A (SEA) and B (SEB) are classical models of superantigens (SAg), which induce potent T-cell-stimulating activity by forming complexes with MHC class II molecules on antigen-presenting cells. This large-scale activation of T-cells is accompanied by increased production of cytokines such as interferon-γ (IFN-γ). Additionally, as we previously reported, IFN-γ-producing CD8(+) T cells act as "helper cells," supporting the ability of dendritic cells to produce interleukin-12 (IL-12)p70. Here, we show that DC pulsed with SAg promote the enhancement of anti-tumor immunity. Murine bone marrow-derived dendritic cells (DC) were pulsed with OVA(257-264) (SIINFEKL), which is an H-2Kb target epitope of EG7 [ovalbumin (OVA)-expressing EL4] cell lines, in the presence of SEA and SEB and were subcutaneously injected into naïve C57BL/6 mice. SAg plus OVA(257-264)-pulsed DC vaccine strongly enhanced peptide-specific CD8(+) T cells exhibiting OVA(257-264)-specific cytotoxic activity and IFN-γ production, leading to the induction of protective immunity against EG7 tumors. Furthermore, cyclophosphamide (CY) added to SAg plus tumor-antigens (OVA(257-264), tumor lysate, or TRP-2) pulsed DC immunization markedly enhanced tumor-specific T-cell expansion and had a significant therapeutic effect against various tumors (EG7, 2LL, and B16). Superantigens are potential candidates for enhancing tumor immunity in DC vaccines.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antineoplastic Agents, Alkylating/therapeutic use
- CD8-Positive T-Lymphocytes
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Cyclophosphamide/therapeutic use
- Cytokines/metabolism
- Dendritic Cells/immunology
- Flow Cytometry
- Histocompatibility Antigens Class II/metabolism
- Interferon-gamma/metabolism
- Interleukin-12/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lymphocyte Activation
- Lymphoma/drug therapy
- Lymphoma/immunology
- Lymphoma/metabolism
- Male
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Mice
- Mice, Inbred C57BL
- Ovalbumin/physiology
- Receptors, G-Protein-Coupled/physiology
- Superantigens/immunology
- Survival Rate
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Tumor Cells, Cultured
- Vaccines, Subunit/therapeutic use
Collapse
Affiliation(s)
- Masato Kato
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Yuichi Ozawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naohiro Seo
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Toshi Nagata
- Department of Health Science, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yukio Koide
- Department of Infectious Diseases, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Pawel Kalinski
- Department of Surgery, Immunology, Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA USA
| | - Hirotoshi Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| | - Kingo Chida
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192 Japan
| |
Collapse
|
34
|
Gene detection of staphylococcal enterotoxins in production strain of staphylococcin injection and superantigenic activity of rSEK and rSEQ. World J Microbiol Biotechnol 2011. [DOI: 10.1007/s11274-011-0779-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
35
|
Mauguen A, Le Deley MC, Zohar S. Dose-finding approach for dose escalation with overdose control considering incomplete observations. Stat Med 2011; 30:1584-94. [DOI: 10.1002/sim.4128] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 09/19/2010] [Indexed: 11/09/2022]
|
36
|
|
37
|
Forsberg G, Skartved NJ, Wallén-Öhman M, Nyhlén HC, Behm K, Hedlund G, Nederman T. Naptumomab Estafenatox, an Engineered Antibody-superantigen Fusion Protein With Low Toxicity and Reduced Antigenicity. J Immunother 2010; 33:492-9. [DOI: 10.1097/cji.0b013e3181d75820] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
38
|
Tighiouart M, Rogatko A. Dose Finding with Escalation with Overdose Control (EWOC) in Cancer Clinical Trials. Stat Sci 2010. [DOI: 10.1214/10-sts333] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
39
|
Robinson MK, Alpaugh RK, Borghaei H. Naptumomab estafenatox: a new immunoconjugate. Expert Opin Biol Ther 2010; 10:273-9. [PMID: 20053143 DOI: 10.1517/14712590903575620] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD New agents that specifically engage the immune system are being tested in a variety of malignancies. This review provides an overview of naptumomab, an immunotoxin, with encouraging clinical activity in Phase I trials. AREAS COVERED IN THIS REVIEW This review examines the preclinical and the published clinical data with regards to naptumomab. WHAT THE READER WILL GAIN This review provides the reader with an understanding of the mechanism of action, immunology, pharmacokinetics and clinical activity of this agent. TAKE HOME MESSAGE Naptumomab has a unique mechanism of action and appears to be an active agent in the treatment of refractory solid tumors such as renal cell carcinoma.
Collapse
Affiliation(s)
- Matthew K Robinson
- Fox Chase Cancer Center, Department of Medical Oncology, 333 Cottman Ave, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|
40
|
Elkord E, Shablak A, Stern PL, Hawkins RE. 5T4 as a target for immunotherapy in renal cell carcinoma. Expert Rev Anticancer Ther 2010; 9:1705-9. [PMID: 19954280 DOI: 10.1586/era.09.152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
41
|
Sundstedt A, Celander M, Öhman MW, Forsberg G, Hedlund G. Immunotherapy with tumor-targeted superantigens (TTS) in combination with docetaxel results in synergistic anti-tumor effects. Int Immunopharmacol 2009; 9:1063-70. [DOI: 10.1016/j.intimp.2009.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 04/24/2009] [Accepted: 04/27/2009] [Indexed: 12/22/2022]
|
42
|
Identification of protein components and quantitative immunoassay for SEC2 in staphylococcin injection. J Pharm Biomed Anal 2009; 50:79-85. [DOI: 10.1016/j.jpba.2009.03.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2008] [Revised: 03/24/2009] [Accepted: 03/25/2009] [Indexed: 11/19/2022]
|
43
|
Borghaei H, Alpaugh K, Hedlund G, Forsberg G, Langer C, Rogatko A, Hawkins R, Dueland S, Lassen U, Cohen RB. Phase I dose escalation, pharmacokinetic and pharmacodynamic study of naptumomab estafenatox alone in patients with advanced cancer and with docetaxel in patients with advanced non-small-cell lung cancer. J Clin Oncol 2009; 27:4116-23. [PMID: 19636016 DOI: 10.1200/jco.2008.20.2515] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Two phase I studies were conducted of ABR-217620 alone or in combination with docetaxel. This is a recombinant fusion protein consisting of a mutated variant of the superantigen staphylococcal enterotoxin E (SEA/E-120) linked to fragment antigen binding moiety of a monoclonal antibody recognizing the tumor-associated antigen 5T4. PATIENTS AND METHODS Patients with non-small-cell lung cancer (NSCLC), pancreatic cancer (PC), and renal cell cancer (RCC) received 5 daily boluses of ABR-217620 (3-month cycles) in escalating doses to determine the maximum-tolerated dose (MTD; ABR-217620 dose escalation monotherapy [MONO] study). Doses were selected based on individual patient anti-SEA/E-120 titers pretreatment. Patients with NSCLC received 4 daily, escalating doses of ABR-217620 followed by docetaxel in 21-day cycles (ABR-217620 dose escalation combination with docetaxel [COMBO] study). RESULTS Thirty-nine patients were enrolled in the MONO study and 13 were enrolled in the COMBO study. The monotherapy MTD was 26 microg/kg (NSCLC and PC) and 15 microg/kg (RCC). Dose-limiting toxicities (DLTs) in the MONO study were fever, hypotension, acute liver toxicity, and vascular leak syndrome. In the COMBO study, the MTD was 22 microg/kg (neutropenic sepsis). Adverse events included grade 1 to 2 fever, hypotension, nausea, and chills. Treatment caused a systemic increase of inflammatory cytokines and selective expansion of SEA/E-120 reactive T-cells. Tumor biopsies demonstrated T-cell infiltration after therapy. Fourteen patients (36%) had stable disease (SD) on day 56 of the MONO study. Two patients (15%) in the COMBO study had partial responses, one in a patient with progressive disease on prior docetaxel, and five patients (38%) had SD on day 56. CONCLUSION ABR-217620 was well tolerated with evidence of immunological activity and antitumor activity.
Collapse
Affiliation(s)
- Hossein Borghaei
- DO, Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wang X, Xu M, Zhang H, Liu J, Li X, Zhang C. Enhancement of superantigen activity and antitumor response of staphylococcal enterotoxin C2 by site-directed mutagenesis. Cancer Immunol Immunother 2009; 58:677-86. [PMID: 18818919 PMCID: PMC11030829 DOI: 10.1007/s00262-008-0590-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 09/03/2008] [Indexed: 10/21/2022]
Abstract
Bacterial superantigen staphylococcal enterotoxins (SEs) tremendously stimulate polyclonal T cells bearing particular TCR Vbeta domains when binding to MHC II molecules, suggesting that they could be a candidate of new antitumor agent. SEC2, an important member of superantigen family, has been used in clinical trial as an immunotherapy agent for cancer treatment in China, and obtained some encouraging effects. However, the presence of immunosuppression and endotoxic activity limits the therapeutic dosage of SEC2, and influences its antitumor effect in clinic. Therefore, the enhancement of superantigen activity and antitumor effect of SEC2 could effectively make compensation for the disadvantages mentioned above. In this study, a superantigen SEC2(T20L/G22E) mutant was generated by site-directed mutagenesis, and efficiently expressed in E. coli BL21(DE3). The results showed that SEC2(T20L/G22E) mutant exhibited a significantly enhanced superantigen activity and antitumor response, compared with native SEC2 in vitro. Further toxicity assay in vivo indicated that SEC2(T20L/G22E) mutant had no significant increase in emetic and pyrogenic activity compared with SEC2, which suggested that the mutant SEC2(T20L/G22E) could be used as a potentially powerful candidate for cancer immunotherapy, and could make compensation for the deficiency of native SEC2 in clinic.
Collapse
Affiliation(s)
- Xiaogang Wang
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
- Graduate School of Chinese Academy of Sciences, 100039 Beijing, China
| | - Mingkai Xu
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| | - Huiwen Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| | - Jie Liu
- China Medical University, 110001 Shenyang, China
| | - Xu Li
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| | - Chenggang Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, 110016 Shenyang, China
| |
Collapse
|
45
|
Chu PL, Lin Y, Shih WJ. Unifying CRM and EWOC designs for phase I cancer clinical trials. J Stat Plan Inference 2009. [DOI: 10.1016/j.jspi.2008.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
46
|
Iasonos A, Wilton AS, Riedel ER, Seshan VE, Spriggs DR. A comprehensive comparison of the continual reassessment method to the standard 3 + 3 dose escalation scheme in Phase I dose-finding studies. Clin Trials 2009; 5:465-77. [PMID: 18827039 PMCID: PMC2637378 DOI: 10.1177/1740774508096474] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background An extensive literature has covered the statistical properties of the Continual Reassessment Method (CRM) and the modifications of this method. While there are some applications of CRM designs in recent Phase I trials, the standard method (SM) of escalating doses after three patients with an option for an additional three patients SM remains very popular, mainly due to its simplicity. From a practical perspective, clinicians are interested in designs that can estimate the MTD using fewer patients for a fixed number of doses, or can test more dose levels for a given sample size. Purpose This article compares CRM-based methods with the SM in terms of the number of patients needed to reach the MTD, total sample size required, and trial duration. Methods The comparisons are performed under two alternative schemes: a fixed or a varying sample approach with the implementation of a stopping rule. The stopping rule halts the trial if the confidence interval around the MTD is within a pre-specified bound. Our simulations evaluated several CRM-based methods under different scenarios by varying the number of dose levels from five to eight and the location of the true MTD. Results CRM and SM are comparable in terms of how fast they reach the MTD and the total sample size required when testing a limited number of dose levels (≤5), but as the number of dose levels increases, CRM reaches the MTD in fewer patients when used with a fixed sample of 20 patients. However, a sample size of 20–25 patients is not sufficient to achieve a narrow precision around the estimated toxicity rate at the MTD. Limitations We focused on methods with practical design features that are of interest to clinicians. However, there are several alternative CRM-based designs that are not investigated in this manuscript, and hence our results are not generalizable to other designs. Conclusions We show that CRM-based methods are an improvement over the SM in terms of accuracy and optimal dose allocation in almost all cases, except when the true dose is among the lower levels.
Collapse
Affiliation(s)
- Alexia Iasonos
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
47
|
Thall PF, Nguyen HQ, Estey EH. Patient-specific dose finding based on bivariate outcomes and covariates. Biometrics 2008; 64:1126-36. [PMID: 18355387 DOI: 10.1111/j.1541-0420.2008.01009.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
SUMMARY A Bayesian sequential dose-finding procedure based on bivariate (efficacy, toxicity) outcomes that accounts for patient covariates and dose-covariate interactions is presented. Historical data are used to obtain an informative prior on covariate main effects, with uninformative priors assumed for all dose effect parameters. Elicited limits on the probabilities of efficacy and toxicity for each of a representative set of covariate vectors are used to construct bounding functions that determine the acceptability of each dose for each patient. Elicited outcome probability pairs that are equally desirable for a reference patient are used to define two different posterior criteria, either of which may be used to select an optimal covariate-specific dose for each patient. Because the dose selection criteria are covariate specific, different patients may receive different doses at the same point in the trial, and the set of eligible patients may change adaptively during the trial. The method is illustrated by a dose-finding trial in acute leukemia, including a simulation study.
Collapse
Affiliation(s)
- Peter F Thall
- Department of Biostatistics, The University of Texas, MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
48
|
|
49
|
Shaw DM, Connolly NB, Patel PM, Kilany S, Hedlund G, Nordle O, Forsberg G, Zweit J, Stern PL, Hawkins RE. A phase II study of a 5T4 oncofoetal antigen tumour-targeted superantigen (ABR-214936) therapy in patients with advanced renal cell carcinoma. Br J Cancer 2007; 96:567-74. [PMID: 17285137 PMCID: PMC2360042 DOI: 10.1038/sj.bjc.6603567] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In a phase II study, 43 renal cell carcinoma patients were treated with individualised doses of ABR-214936; a fusion of a Fab recognising the antigen 5T4, and Staphylococcal enterotoxin A. Drug was given intravenously on 4 consecutive days, treatment was repeated 1 month later. Treatment was associated with moderate fever and nausea, but well tolerated. Of 40 evaluable patients, 28 had disease control at 2 months, and at 4 months, one patient showed partial response (PR) and 16 patients stable disease. Median survival, with minimum follow-up of 26 months was 19.7 months with 13 patients alive to date. Stratification by the Motzer's prognostic criteria highlights prolonged survival compared to published expectation. Patients receiving higher drug exposure had greater disease control and lived almost twice as long as expected, whereas the low-exposure patients survived as expected. Sustained interleukin-2 (IL-2) production after a repeated injection appears to be a biomarker for clinical effect, as the induced-IL-2 level on the day 2 of treatment correlated with survival. The high degree of disease control and the prolonged survival suggest that this treatment can be effective. These findings will be used in the trial design for the next generation of drug, with reduced antigenicity and toxicity.
Collapse
Affiliation(s)
- D M Shaw
- Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Si S, Sun Y, Li Z, Ge W, Zhang X, Hu P, Huang Y, Chen G, Song H, Huang Y, Ma B, Li X, Sui Y. Gene therapy by membrane-expressed superantigen for α-fetoprotein-producing hepatocellular carcinoma. Gene Ther 2006; 13:1603-10. [PMID: 16855617 DOI: 10.1038/sj.gt.3302823] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Staphylococcus enterotoxin A (SEA) is a powerful immunostimulant, which can stimulate T cells bearing certain T-cell receptor beta-chain variable regions, when bound to major histocompatibility complex II molecules. In vivo administration of intact superantigen in sufficient therapeutic amounts risks unwanted cytotoxicity against normal cells. In this study, we used SEA fused with CD80 transmembrane region (named as SEAtm) driven by alpha-fetoprotein (AFP) enhancer/promoter to reduce toxicity and to improve safety and efficiency in the application of SEA. We demonstrated that SEAtm by adenovirus from the AFP enhancer/promoter (AdAFPSEA) could be expressed on the surface of AFP-producing cell line Hepa1-6 instead of non-AFP-producing cell lines. Hepa1-6 infected by recombinant adenovirus stimulated proliferation of splenocytes and activated CD4(+) and CD8(+) T cells in vitro. After AdAFPSEA was injected into the subcutaneously established hepatoma in vivo, the expression of SEA was detected in tumor tissues, which subsequently induced tumor-specific cytotoxic T cells in spleen. Moreover, hepatocellular carcinoma (HCC) xenografts were suppressed by treatment with AdAFPSEA and the survival time of treated mice was prolonged. These findings suggest that membrane-expressed SEA by adenovirus from AdAFPSEA can generate stronger local and systemic antitumor responses against HCC.
Collapse
Affiliation(s)
- S Si
- State Key Laboratory of Cancer Biology, Department of Pathology, XiJing Hospital, Fourth Military Medmedical University, Shanxi Province, Xi'an, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|