1
|
Thein KZ, Myat YM, Park BS, Panigrahi K, Kummar S. Target-Driven Tissue-Agnostic Drug Approvals-A New Path of Drug Development. Cancers (Basel) 2024; 16:2529. [PMID: 39061168 PMCID: PMC11274498 DOI: 10.3390/cancers16142529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The regulatory approvals of tumor-agnostic therapies have led to the re-evaluation of the drug development process. The conventional models of drug development are histology-based. On the other hand, the tumor-agnostic drug development of a new drug (or combination) focuses on targeting a common genomic biomarker in multiple cancers, regardless of histology. The basket-like clinical trials with multiple cohorts allow clinicians to evaluate pan-cancer efficacy and toxicity. There are currently eight tumor agnostic approvals granted by the Food and Drug Administration (FDA). This includes two immune checkpoint inhibitors, and five targeted therapy agents. Pembrolizumab is an anti-programmed cell death protein-1 (PD-1) antibody that was the first FDA-approved tumor-agnostic treatment for unresectable or metastatic microsatellite instability-high (MSI-H) or deficient mismatch repair (dMMR) solid tumors in 2017. It was later approved for tumor mutational burden-high (TMB-H) solid tumors, although the TMB cut-off used is still debated. Subsequently, in 2021, another anti-PD-1 antibody, dostarlimab, was also approved for dMMR solid tumors in the refractory setting. Patients with fusion-positive cancers are typically difficult to treat due to their rare prevalence and distribution. Gene rearrangements or fusions are present in a variety of tumors. Neurotrophic tyrosine kinase (NTRK) fusions are present in a range of pediatric and adult solid tumors in varying frequency. Larotrectinib and entrectinib were approved for neurotrophic tyrosine kinase (NTRK) fusion-positive cancers. Similarly, selpercatinib was approved for rearranged during transfection (RET) fusion-positive solid tumors. The FDA approved the first combination therapy of dabrafenib, a B-Raf proto-oncogene serine/threonine kinase (BRAF) inhibitor, plus trametinib, a mitogen-activated protein kinase (MEK) inhibitor for patients 6 months or older with unresectable or metastatic tumors (except colorectal cancer) carrying a BRAFV600E mutation. The most recent FDA tumor-agnostic approval is of fam-trastuzumab deruxtecan-nxki (T-Dxd) for HER2-positive solid tumors. It is important to identify and expeditiously develop drugs that have the potential to provide clinical benefit across tumor types.
Collapse
Affiliation(s)
- Kyaw Z. Thein
- Division of Hematology and Medical Oncology, Comprehensive Cancer Centers of Nevada—Central Valley, 3730 S Eastern Ave, Las Vegas, NV 89169, USA
- Department of Medicine, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), 4505 S, Maryland Pkwy, Las Vegas, NV 89154, USA
- College of Osteopathic Medicine, Touro University Nevada, Touro College and University System, 874 American Pacific Dr, Henderson, NV 89014, USA
| | - Yin M. Myat
- Belfield Campus, University College Dublin (UCD) School of Medicine, D04 V1W8 Dublin, Ireland;
- Department of Internal Medicine, One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA;
| | - Byung S. Park
- OHSU-PSU School of Public Health, Portland, OR 97201, USA;
- Biostatistics Shared Resource, OHSU Knight Cancer Institute, OHSU School of Medicine, Portland, OR 97239, USA
| | - Kalpana Panigrahi
- Department of Internal Medicine, One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA;
| | - Shivaani Kummar
- Division of Hematology & Medical Oncology, Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA;
| |
Collapse
|
2
|
Lavacchi D, Fancelli S, Buttitta E, Vannini G, Guidolin A, Winchler C, Caliman E, Vannini A, Giommoni E, Brugia M, Cianchi F, Pillozzi S, Roviello G, Antonuzzo L. Perioperative Tailored Treatments for Gastric Cancer: Times Are Changing. Int J Mol Sci 2023; 24:4877. [PMID: 36902306 PMCID: PMC10003389 DOI: 10.3390/ijms24054877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Resectable gastric or gastroesophageal (G/GEJ) cancer is a heterogeneous disease with no defined molecularly based treatment strategy. Unfortunately, nearly half of patients experience disease recurrence despite standard treatments (neoadjuvant and/or adjuvant chemotherapy/chemoradiotherapy and surgery). In this review, we summarize the evidence of potential tailored approaches in perioperative treatment of G/GEJ cancer, with a special focus on patients with human epidermal growth factor receptor-2(HER2)-positive and microsatellite instability-high (MSI-H) tumors. In patients with resectable MSI-H G/GEJ adenocarcinoma, the ongoing INFINITY trial introduces the concept of non-operative management for patients with complete clinical-pathological-molecular response, and this could be a novel and potential practice changing strategy. Other pathways involving vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), claudin18 isoform 2 (CLDN18.2), and DNA damage repair proteins are also described, with limited evidence until now. Although tailored therapy appears to be a promising strategy for resectable G/GEJ cancer, there are several methodological issues to address: inadequate sample size for pivotal trials, underestimation of subgroup effects, and choice of primary endpoint (tumor-centered vs. patient-centered endpoints). A better optimization of G/GEJ cancer treatment allows maximizing patient outcomes. In the perioperative phase, although caution is mandatory, times are changing and tailored strategies could introduce new treatment concepts. Overall, MSI-H G/GEJ cancer patients possess the characteristics to be the subgroup that could receive the most benefit from a tailored approach.
Collapse
Affiliation(s)
- Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Sara Fancelli
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Eleonora Buttitta
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Gianmarco Vannini
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Alessia Guidolin
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Costanza Winchler
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Agnese Vannini
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Marco Brugia
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Unit of Digestive Surgery, Careggi University Hospital, 50134 Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | | | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| |
Collapse
|
3
|
Conroy JM, Pabla S, Glenn ST, Seager RJ, Van Roey E, Gao S, Burgher B, Andreas J, Giamo V, Mallon M, Lee YH, DePietro P, Nesline M, Wang Y, Lenzo FL, Klein R, Zhang S. A scalable high-throughput targeted next-generation sequencing assay for comprehensive genomic profiling of solid tumors. PLoS One 2021; 16:e0260089. [PMID: 34855780 PMCID: PMC8639101 DOI: 10.1371/journal.pone.0260089] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/03/2021] [Indexed: 11/18/2022] Open
Abstract
Timely and accurate identification of molecular alterations in solid tumors is essential for proper management of patients with advanced cancers. This has created a need for rapid, scalable comprehensive genomic profiling (CGP) systems that detect an increasing number of therapeutically-relevant variant types and molecular signatures. In this study, we assessed the analytical performance of the TruSight Oncology 500 High-Throughput assay for detection of somatic alterations from formalin-fixed paraffin-embedded tissue specimens. In parallel, we developed supporting software and automated sample preparation systems designed to process up to 70 clinical samples in a single NovaSeq 6000TM sequencing run with a turnaround time of <7 days from specimen receipt to report. The results demonstrate that the scalable assay accurately and reproducibly detects small variants, copy number alterations, microsatellite instability (MSI) and tumor mutational burden (TMB) from 40ng DNA, and multiple gene fusions, including known and unknown partners and splice variants from 20ng RNA. 717 tumor samples and reference materials with previously known alterations in 96 cancer-related genes were sequenced to evaluate assay performance. All variant classes were reliably detected at consistent and reportable variant allele percentages with >99% overall accuracy and precision. Our results demonstrate that the high-throughput CGP assay is a reliable method for accurate detection of molecular alterations in support of precision therapeutics in oncology. The supporting systems and scalable workflow allow for efficient interpretation and prompt reporting of hundreds of patient cancer genomes per week with excellent analytical performance.
Collapse
Affiliation(s)
- Jeffrey M. Conroy
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
- Research Support Services, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - Sarabjot Pabla
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Sean T. Glenn
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
- Laboratory Operations, OmniSeq Inc., Buffalo, New York, United States of America
- HemePath Molecular, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - R. J. Seager
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Erik Van Roey
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Shuang Gao
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Blake Burgher
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Jonathan Andreas
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Vincent Giamo
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Melissa Mallon
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Yong Hee Lee
- Clinical Evidence Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Paul DePietro
- Clinical Evidence Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Mary Nesline
- Clinical Evidence Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Yirong Wang
- Information Technology, OmniSeq Inc., Buffalo, New York, United States of America
| | - Felicia L. Lenzo
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Roger Klein
- Medical Affairs, OmniSeq Inc., Buffalo, New York, United States of America
| | - Shengle Zhang
- Laboratory Operations, OmniSeq Inc., Buffalo, New York, United States of America
| |
Collapse
|
4
|
Cambrosio A, Campbell J, Keating P, Polk JB, Aguilar-Mahecha A, Basik M. Healthcare policy by other means: Cancer clinical research as "oncopolicy". Soc Sci Med 2021; 292:114576. [PMID: 34826765 DOI: 10.1016/j.socscimed.2021.114576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 11/29/2022]
Abstract
Social studies of biomedicine often focus on how exogenous policies shape the medical domain. While policy agendas no doubt affect complex biomedical projects, in the present paper we analyze a different dynamic, namely how oncologists enact policy as part of several flagship precision oncology endeavors. Empirically, the article focuses on the U.S. TAPUR trial, the Dutch DRUP trial, and the Canadian CAPTUR trial, which have recently been joined by similar Scandinavian studies. Taken together, these trials represent innovative forms of clinical research that, beyond their varying experimental nature, have been designed to transform the evidential processes to provide access to biomarker-driven treatments. Along with gathering evidence on effectiveness of off-label targeted therapies, their explicit goals include the recentering of a major professional organization around research, and the reframing of healthcare as a learning system seamlessly connecting epistemic, organizational, and economic issues. Accordingly, we analyze the design and implementation of these trials as a form of (onco)policy by other means.
Collapse
Affiliation(s)
- Alberto Cambrosio
- Department of Social Studies of Medicine, McGill University, Montreal, Canada.
| | - Jonah Campbell
- Department of Social Studies of Medicine, McGill University, Montreal, Canada
| | - Peter Keating
- Department of History, Université du Québec à Montréal, Montreal, Canada
| | - Jessica B Polk
- Department of Social Studies of Medicine, McGill University, Montreal, Canada
| | | | - Mark Basik
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
5
|
Basket trials: From tumour gnostic to tumour agnostic drug development. Cancer Treat Rev 2020; 90:102082. [DOI: 10.1016/j.ctrv.2020.102082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
|
6
|
Pederzoli F, Bandini M, Marandino L, Ali SM, Madison R, Chung J, Ross JS, Necchi A. Targetable gene fusions and aberrations in genitourinary oncology. Nat Rev Urol 2020; 17:613-625. [PMID: 33046892 DOI: 10.1038/s41585-020-00379-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
Gene fusions result from either structural chromosomal rearrangement or aberrations caused by splicing or transcriptional readthrough. The precise and distinctive presence of fusion genes in neoplastic tissues and their involvement in multiple pathways central to cancer development, growth and survival make them promising targets for personalized therapy. In genitourinary malignancies, rearrangements involving the E26 transformation-specific family of transcription factors have emerged as very frequent alterations in prostate cancer, especially the TMPRSS2-ERG fusion. In renal malignancies, Xp11 and t(6;11) translocations are hallmarks of a distinct pathological group of tumours described as microphthalmia-associated transcription factor family translocation-associated renal cell carcinomas. Novel druggable fusion events have been recognized in genitourinary malignancies, leading to the activation of several clinical trials. For instance, ALK-rearranged renal cell carcinomas have shown responses to alectinib and crizotinib. Erdafitinib has been tested for the treatment of FGFR-rearranged bladder cancer. Other anti-fibroblast growth factor receptor 3 (FGFR3) compounds are showing promising results in the treatment of bladder cancer, including infigratinib and pemigatinib, and all are currently in clinical trials.
Collapse
Affiliation(s)
- Filippo Pederzoli
- Urological Research Institute (URI), Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy.
| | - Marco Bandini
- Urological Research Institute (URI), Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Laura Marandino
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Siraj M Ali
- Foundation Medicine Inc., Cambridge, MA, USA
| | | | - Jon Chung
- Foundation Medicine Inc., Cambridge, MA, USA
| | - Jeffrey S Ross
- Foundation Medicine Inc., Cambridge, MA, USA.,Upstate Medical University, Syracuse, NY, USA
| | - Andrea Necchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
7
|
Bitterman DS, Cagney DN, Singer LL, Nguyen PL, Catalano PJ, Mak RH. Master Protocol Trial Design for Efficient and Rational Evaluation of Novel Therapeutic Oncology Devices. J Natl Cancer Inst 2020; 112:229-237. [PMID: 31504680 PMCID: PMC7073911 DOI: 10.1093/jnci/djz167] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/26/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022] Open
Abstract
Historically, the gold standard for evaluation of cancer therapeutics, including medical devices, has been the randomized clinical trial. Although high-quality clinical data are essential for safe and judicious use of therapeutic oncology devices, class II devices require only preclinical data for US Food and Drug Administration approval and are often not rigorously evaluated prior to widespread uptake. Herein, we review master protocol design in medical oncology and its application to therapeutic oncology devices, using examples from radiation oncology. Unique challenges of clinical testing of radiation oncology devices (RODs) include patient and treatment heterogeneity, lack of funding for trials by industry and health-care payers, and operator dependence. To address these challenges, we propose the use of master protocols to optimize regulatory, financial, administrative, quality assurance, and statistical efficiency of trials evaluating RODs. These device-specific master protocols can be extrapolated to other devices and encompass multiple substudies with the same design, statistical considerations, logistics, and infrastructure. As a practical example, we outline our phase I and II master protocol trial of stereotactic magnetic resonance imaging–guided adaptive radiotherapy, which to the best of our knowledge is the first master protocol trial to test a ROD. Development of more efficient clinical trials is needed to promote thorough evaluation of therapeutic oncology devices, including RODs, in a resource-limited environment, allowing more practical and rapid identification of the most valuable advances in our field.
Collapse
Affiliation(s)
- Danielle S Bitterman
- Harvard Radiation Oncology Program, Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA.,Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Daniel N Cagney
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lisa L Singer
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Paul L Nguyen
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Paul J Catalano
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Raymond H Mak
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
8
|
Lowder CY, Dhir T, Goetz AB, Thomsett HL, Bender J, Tatarian T, Madhavan S, Petricoin EF, Blais E, Lavu H, Winter JM, Posey J, Brody JR, Pishvaian MJ, Yeo CJ. A step towards personalizing next line therapy for resected pancreatic and related cancer patients: A single institution's experience. Surg Oncol 2020; 33:118-125. [PMID: 32561076 PMCID: PMC7498307 DOI: 10.1016/j.suronc.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/09/2019] [Accepted: 02/06/2020] [Indexed: 12/17/2022]
Abstract
Background: There is a lack of precision medicine in pancreatic ductal adenocarcinoma (PDA) and related cancers, and outcomes for patients with this diagnosis remain poor despite decades of research investigating this disease. Therefore, it is necessary to explore novel therapeutic options for these patients who may benefit from personalized therapies. Objective: Molecular profiling of hepatopancreaticobiliary malignancies at our institution, including but not limited to PDA, was initiated to assess the feasibility of incorporating molecular profiling results into patient oncological therapy planning. Methods: All eligible patients from Thomas Jefferson University (TJU) with hepatopancreaticobiliary tumors including PDA, who agreed to molecular testing profiling, were prospectively enrolled in a registry study from December 2014 to September 2017 and their tumor samples were tested to identify molecular markers that can be used to guide therapy options in the future. Next generation sequencing (NGS) and protein expression in tumor samples were tested at CLIA-certified laboratories. Prospective clinicopathologic data were extracted from medical records and compiled in a de-identified fashion. Results: Seventy eight (78) patients were enrolled in the study, which included 65/78 patients with PDA (local and metastatic) and out of that subset, 52/65 patients had surgically resected PDA. Therapy recommendations were generated based on molecular and clinicopathologic data for all enrolled patients. NGS uncovered actionable alterations in 25/52 surgically resected PDAs (48%) which could be used to guide therapy options in the future. High expression of three proteins, TS (p ¼ 0.005), ERCC1 (p = 0.001), and PD-1 (p = 0.04), was associated with reduced recurrence-free survival (RFS), while TP53 mutations were correlated with longer RFS (p = 0.01). Conclusions: The goal of this study was to implement a stepwise strategy to identify and profile resected PDAs at our institution. Consistent with previous studies, approximately half of patients with resected PDA harbor actionable mutations with possible targeted therapeutic implications. Ongoing studies will determine the clinical value of identifying these mutations in patients with resected PDA.
Collapse
Affiliation(s)
- Cinthya Y Lowder
- Department of Surgery, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Teena Dhir
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Austin B Goetz
- Department of Surgery, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Henry L Thomsett
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Talar Tatarian
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Subha Madhavan
- Perthera, Inc, McLean, VA, USA; The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Harish Lavu
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jordan M Winter
- University Hospital Seidman Cancer Center, Cleveland, OH, USA; University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - James Posey
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jonathan R Brody
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael J Pishvaian
- Perthera, Inc, McLean, VA, USA; The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Charles J Yeo
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Raheem F, Kim P, Grove M, Kiel PJ. Precision Genomic Practice in Oncology: Pharmacist Role and Experience in an Ambulatory Care Clinic. PHARMACY 2020; 8:pharmacy8010032. [PMID: 32182657 PMCID: PMC7151676 DOI: 10.3390/pharmacy8010032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022] Open
Abstract
Recent advancements in molecular testing, the availability of cost-effective technology, and novel approaches to clinical trial design have facilitated the implementation of tumor genome sequencing into standard of care oncology practices. Current models of precision oncology practice include specialized clinics or consultation services based on a molecular tumor board (MTB) approach. MTBs are comprised of interprofessional teams of clinicians and scientists who evaluate tumors at the molecular level to guide patient-specific targeted therapy. The practice of precision oncology utilizing MTB-based models is an emerging approach, transforming precision genomics from a novel concept into clinical practice. This rapid shift in practice from cytotoxic therapy to targeted medicine poses challenges, yet brings exciting opportunities to clinical pharmacists practicing in hematology and oncology. Only a few precision genomics programs in the United States have a strong pharmacy presence with oncology pharmacists serving in leadership roles in research, interpreting genomic sequencing, making treatment recommendations, and facilitating off-label drug procurement. This article describes the experience of the precision medicine clinic at the Indiana University Health Simon Cancer Center, with emphasis on the role of the pharmacist in the precision oncology initiative.
Collapse
|
10
|
Chung C. Driving toward precision medicine for B cell lymphomas: Targeting the molecular pathogenesis at the gene level. J Oncol Pharm Pract 2020; 26:943-966. [DOI: 10.1177/1078155219895079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lymphomas are a diverse group of hematologic malignancies that arise from either T cell, B cell or the natural killer cell lineage. B cell lymphomas arise from gene mutations with critical functions during normal B cell development. Recent advances in the understanding of molecular pathogenesis demonstrate that many different recurrent genomic and molecular abnormalities and dysregulated oncogenic regulatory pathways exist for many subtypes of B cell lymphomas, both across and within histological subtypes. Pathogenetic processes such as (1) chromosomal aberrations, for example, t(14;18) in follicular lymphoma, t(11;14) in mantle cell lymphoma, t(8;14) in Burkitt lymphoma; dysregulations in signaling pathways of (2) nuclear factor- κB (NF-κB); (3) B cell receptor (BCR); (4) Janus kinase/signal transducers and transcription activators (JAK-STAT); (5) impaired apoptosis/cell cycle regulation due to mutated, rearranged or amplified MYC, BCL-2, BCL-6 proto-oncogenes; (6) epigenetic aberrations may contribute to pathogenesis. More studies are under way to elucidate the molecular heterogeneity underlying many types of lymphomas that account for variable responses to treatment, generation of subclones and treatment resistance. Although significant research is still needed, targeted therapy promises to provide new options for the treatment of patients with lymphomas. This article provides a non-exhaustive overview on the current understanding on the genetics of pathogenesis of B cell lymphomas and their therapeutic implications.
Collapse
Affiliation(s)
- Clement Chung
- Houston Methodist Baytown Hospital, Baytown, TX, USA
| |
Collapse
|
11
|
Li W, Zhao J, Li X, Chen C, Beckman RA. Multi‐stage enrichment and basket trial designs with population selection. Stat Med 2019; 38:5470-5485. [DOI: 10.1002/sim.8371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/03/2019] [Accepted: 08/16/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Wen Li
- Biostatistics and Research Decision Sciences, Merck Research LaboratoriesMerck & Co, Inc Kenilworth New Jersey
| | - Jing Zhao
- Biostatistics and Research Decision Sciences, Merck Research LaboratoriesMerck & Co, Inc Kenilworth New Jersey
| | - Xiaoyun Li
- Biostatistics and Research Decision Sciences, Merck Research LaboratoriesMerck & Co, Inc Kenilworth New Jersey
| | - Cong Chen
- Biostatistics and Research Decision Sciences, Merck Research LaboratoriesMerck & Co, Inc Kenilworth New Jersey
| | - Robert A. Beckman
- Departments of Oncology and of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical InformaticsGeorgetown University Medical Center Washington District of Columbia
| |
Collapse
|
12
|
van der Velden DL, Hoes LR, van der Wijngaart H, van Berge Henegouwen JM, van Werkhoven E, Roepman P, Schilsky RL, de Leng WWJ, Huitema ADR, Nuijen B, Nederlof PM, van Herpen CML, de Groot DJA, Devriese LA, Hoeben A, de Jonge MJA, Chalabi M, Smit EF, de Langen AJ, Mehra N, Labots M, Kapiteijn E, Sleijfer S, Cuppen E, Verheul HMW, Gelderblom H, Voest EE. The Drug Rediscovery protocol facilitates the expanded use of existing anticancer drugs. Nature 2019; 574:127-131. [DOI: 10.1038/s41586-019-1600-x] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 08/14/2019] [Indexed: 12/19/2022]
|
13
|
Dehghani M, Rosenblatt KP, Li L, Rakhade M, Amato RJ. Validation and Clinical Applications of a Comprehensive Next Generation Sequencing System for Molecular Characterization of Solid Cancer Tissues. Front Mol Biosci 2019; 6:82. [PMID: 31681791 PMCID: PMC6798036 DOI: 10.3389/fmolb.2019.00082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/23/2019] [Indexed: 12/18/2022] Open
Abstract
Identification of somatic molecular alterations in primary and metastatic solid tumor specimens can provide critical information regarding tumor biology and its heterogeneity, and enables the detection of molecular markers for clinical personalized treatment assignment. However, the optimal methods and target genes for clinical use are still being in development. Toward this end, we validated a targeted amplification-based NGS panel (Oncomine comprehensive assay v1) on a personal genome machine sequencer for molecular profiling of solid tumors. This panel covers 143 genes, and requires low amounts of DNA (20 ng) and RNA (10 ng). We used 27 FFPE tissue specimens, 10 cell lines, and 24 commercial reference materials to evaluate the performance characteristics of this assay. We also evaluated the performance of the assay on 26 OCT-embedded fresh frozen specimens (OEFF). The assay was found to be highly specific (>99%) and sensitive (>99%), with low false-positive and false-negative rates for single-nucleotide variants, indels, copy number alterations, and gene fusions. Our results indicate that this is a reliable method to determine molecular alterations in both fixed and fresh frozen solid tumor samples, including core needle biopsies.
Collapse
Affiliation(s)
- Mehdi Dehghani
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States
| | - Kevin P Rosenblatt
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States.,NX Prenatal, Inc., Bellaire, TX, United States.,Consultative Genomics, PLLC, Bellaire, TX, United States
| | - Lei Li
- Principle Health Systems, Houston, TX, United States
| | - Mrudula Rakhade
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States
| | - Robert J Amato
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States
| |
Collapse
|
14
|
Zhang X, Liang Z, Wang S, Lu S, Song Y, Cheng Y, Ying J, Liu W, Hou Y, Li Y, Liu Y, Hou J, Liu X, Shao J, Tai Y, Wang Z, Fu L, Li H, Zhou X, Bai H, Wang M, Lu Y, Yang J, Zhong W, Zhou Q, Yang X, Wang J, Huang C, Liu X, Zhou X, Zhang S, Tian H, Chen Y, Ren R, Liao N, Wu C, Zhu Z, Pan H, Gu Y, Wang L, Liu Y, Zhang S, Liu T, Chen G, Shao Z, Xu B, Zhang Q, Xu R, Shen L, Wu Y, Tumor Biomarker Committee OBOCSOCO(CSCO. Application of next-generation sequencing technology to precision medicine in cancer: joint consensus of the Tumor Biomarker Committee of the Chinese Society of Clinical Oncology. Cancer Biol Med 2019; 16:189-204. [PMID: 31119060 PMCID: PMC6528448 DOI: 10.20892/j.issn.2095-3941.2018.0142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023] Open
Abstract
Next-generation sequencing (NGS) technology is capable of sequencing millions or billions of DNA molecules simultaneously. Therefore, it represents a promising tool for the analysis of molecular targets for the initial diagnosis of disease, monitoring of disease progression, and identifying the mechanism of drug resistance. On behalf of the Tumor Biomarker Committee of the Chinese Society of Clinical Oncology (CSCO) and the China Actionable Genome Consortium (CAGC), the present expert group hereby proposes advisory guidelines on clinical applications of NGS technology for the analysis of cancer driver genes for precision cancer therapy. This group comprises an assembly of laboratory cancer geneticists, clinical oncologists, bioinformaticians, pathologists, and other professionals. After multiple rounds of discussions and revisions, the expert group has reached a preliminary consensus on the need of NGS in clinical diagnosis, its regulation, and compliance standards in clinical sample collection. Moreover, it has prepared NGS criteria, the sequencing standard operation procedure (SOP), data analysis, report, and NGS platform certification and validation.
Collapse
Affiliation(s)
- Xuchao Zhang
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
| | - Shengyue Wang
- National Research Center for Translational Medicine, Shanghai, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Shun Lu
- Lung Tumor Clinical Medical Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yong Song
- Division of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210029, China
| | - Ying Cheng
- Department of Oncology, Jilin Cancer Hospital, Changchun 132002, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Yangqiu Li
- Department of Hematology, First Affiliated Hospital, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 519000, China
| | - Yi Liu
- Laboratory of Oncology, Affiliated Hospital of the Academy of Military Medical Sciences, Beijing 100071, China
| | - Jun Hou
- Department of Oncology, First Clinical College of South China University of Technology/Guangdong Lung Cancer Institute, Guangzhou 510060, China
| | - Xiufeng Liu
- People's Liberation Army Cancer Center of Bayi Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Jianyong Shao
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 519000, China
| | - Yanhong Tai
- Department of Pathology, Affiliated Hospital of the Academy of Military Medical Sciences, Beijing 100071, China
| | - Zheng Wang
- Department of Pathology, Beijing Hospital, Beijing 100071, China
| | - Li Fu
- Department of Breast Cancer Pathology and Research Laboratory of Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hui Li
- Department of Oncology, Jilin Cancer Hospital, Changchun 132002, China
| | - Xiaojun Zhou
- Department of Pathology, Jinling Hospital Nanjing University School of Medicine, Nanjing 210029, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Mengzhao Wang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100006, China
| | - You Lu
- Department of Oncology, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Jinji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xuening Yang
- Guangdong Lung Cancer Institute, Guangdong Provincical Prople's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jie Wang
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Cheng Huang
- Department of Thoracic Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350001, China
| | - Xiaoqing Liu
- Department of Oncology, Affiliated Hospital of the Academy of Military Medical Sciences, Beijing 100071, China
| | - Xiaoyan Zhou
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai 200433, China
| | - Shirong Zhang
- Center for Translational Medicine, Hangzhou First People's Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Hongxia Tian
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | - Yu Chen
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | - Ruibao Ren
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Ning Liao
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangzhou 510080, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200240, China
| | - Zhongzheng Zhu
- Department of Oncology, No. 113 Hospital of People's Liberation Army, Ningbo 315040, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - Liwei Wang
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110016, China
| | - Suzhan Zhang
- Department of Oncology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Tianshu Liu
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Gong Chen
- Department of Colorectal, Sun Yat-sen University Cancer Center, Guangzhou 519000, China
| | - Zhimin Shao
- Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai 200433, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Qingyuan Zhang
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin 150030, China
| | - Ruihua Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 519000, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Medical Research Center, Cancer Center of Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Affiliated Guangdong Provincial People's Hospital, South China University of Technology, Guangzhou 510630, China
| | | |
Collapse
|
15
|
Chen N, Lee JJ. Bayesian hierarchical classification and information sharing for clinical trials with subgroups and binary outcomes. Biom J 2018; 61:1219-1231. [PMID: 30506747 DOI: 10.1002/bimj.201700275] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 11/07/2022]
Abstract
Bayesian hierarchical models have been applied in clinical trials to allow for information sharing across subgroups. Traditional Bayesian hierarchical models do not have subgroup classifications; thus, information is shared across all subgroups. When the difference between subgroups is large, it suggests that the subgroups belong to different clusters. In that case, placing all subgroups in one pool and borrowing information across all subgroups can result in substantial bias for the subgroups with strong borrowing, or a lack of efficiency gain with weak borrowing. To resolve this difficulty, we propose a hierarchical Bayesian classification and information sharing (BaCIS) model for the design of multigroup phase II clinical trials with binary outcomes. We introduce subgroup classification into the hierarchical model. Subgroups are classified into two clusters on the basis of their outcomes mimicking the hypothesis testing framework. Subsequently, information sharing takes place within subgroups in the same cluster, rather than across all subgroups. This method can be applied to the design and analysis of multigroup clinical trials with binary outcomes. Compared to the traditional hierarchical models, better operating characteristics are obtained with the BaCIS model under various scenarios.
Collapse
Affiliation(s)
- Nan Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Offin M, Liu D, Drilon A. Tumor-Agnostic Drug Development. Am Soc Clin Oncol Educ Book 2018; 38:184-187. [PMID: 30231313 DOI: 10.1200/edbk_200831] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Therapies designed to target cancers that harbor specific molecular signatures have reshaped the landscape of oncologic drug development, and advances in next generation sequencing have led to an increase in the identification of these alterations across tumor types. Tumor-agnostic trial designs, such as the "basket trial," have been developed as an approach to study the efficacy of these treatments and increase patient access, especially for patients whose tumors carry these alterations infrequently. We review key aspects of these genomically enriched trial strategies and their impact on drug development and approval.
Collapse
Affiliation(s)
- Michael Offin
- From the Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY; Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dazhi Liu
- From the Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY; Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander Drilon
- From the Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY; Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
17
|
Melloni GE, Guida A, Curigliano G, Botteri E, Esposito A, Kamal M, Le Tourneau C, Riva L, Magi A, de Maria R, Pelicci P, Mazzarella L. Precision Trial Drawer, a Computational Tool to Assist Planning of Genomics-Driven Trials in Oncology. JCO Precis Oncol 2018; 2:1-16. [DOI: 10.1200/po.18.00015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Trials that accrue participants on the basis of genetic biomarkers are a powerful means of testing targeted drugs, but they are often complicated by the rarity of the biomarker-positive population. Umbrella trials circumvent this by testing multiple hypotheses to maximize accrual. However, bigger trials have higher chances of conflicting treatment allocations because of the coexistence of multiple actionable alterations; allocation strategies greatly affect the efficiency of enrollment and should be carefully planned on the basis of relative mutation frequencies, leveraging information from large sequencing projects. Methods We developed software named Precision Trial Drawer (PTD) to estimate parameters that are useful for designing precision trials, most importantly, the number of patients needed to molecularly screen (NNMS) and the allocation rule that maximizes patient accrual on the basis of mutation frequency, systematically assigning patients with conflicting allocations to the drug associated with the rarer mutation. We used data from The Cancer Genome Atlas to show their potential in a 10-arm imaginary trial of multiple cancers on the basis of genetic alterations suggested by the past Molecular Analysis for Personalised Therapy (MAP) conference. We validated PTD predictions versus real data from the SHIVA (A Randomized Phase II Trial Comparing Therapy Based on Tumor Molecular Profiling Versus Conventional Therapy in Patients With Refractory Cancer) trial. Results In the MAP imaginary trial, PTD-optimized allocation reduces number of patients needed to molecularly screen by up to 71.8% (3.5 times) compared with nonoptimal trial designs. In the SHIVA trial, PTD correctly predicted the fraction of patients with actionable alterations (33.51% [95% CI, 29.4% to 37.6%] in imaginary v 32.92% [95% CI, 28.2% to 37.6%] expected) and allocation to specific treatment groups (RAS/MEK, PI3K/mTOR, or both). Conclusion PTD correctly predicts crucial parameters for the design of multiarm genetic biomarker-driven trials. PTD is available as a package in the R programming language and as an open-access Web-based app. It represents a useful resource for the community of precision oncology trialists. The Web-based app is available at https://gmelloni.github.io/ptd/shinyapp.html .
Collapse
Affiliation(s)
- Giorgio E.M. Melloni
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Alessandro Guida
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Giuseppe Curigliano
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Edoardo Botteri
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Angela Esposito
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Maude Kamal
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Christoph Le Tourneau
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Laura Riva
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Alberto Magi
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Ruggero de Maria
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Piergiuseppe Pelicci
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| | - Luca Mazzarella
- Giorgio E.M. Melloni, Harvard Medical School, Boston, MA; Giorgio E.M. Melloni and Laura Riva, Italian Institute of Technology; Alessandro Guida, Giuseppe Curigliano, Angela Esposito, Piergiuseppe Pelicci, and Luca Mazzarella, European Institute of Oncology; Giuseppe Curigliano and Piergiuseppe Pelicci, University of Milan, Milan; Alberto Magi, University of Florence, Florence; Ruggero de Maria, Catholic University, Rome, Italy; Edoardo Botteri, Norwegian Tumor Registry, Oslo, Norway; and Maude Kamal and
| |
Collapse
|
18
|
Valentino F, Borra G, Allione P, Rossi L. Emerging targets in advanced non-small-cell lung cancer. Future Oncol 2018; 14:61-72. [PMID: 29989453 DOI: 10.2217/fon-2018-0099] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
New therapeutic options in non-small-cell lung cancer have been available through a great in-depth and genomic research, improving preclinical disease patterns and identifying the specific toxicity of target therapy. The multidisciplinary approach, increasingly practiced among clinicians, researchers, pharmaceutical companies and ethics committees has allowed the emergence of a new generation of translational clinical trials and the adoption of new technologies (e.g., point-of-care sequencing), then speeding up the development and trade of these new drugs. Consequently, there is a long list of therapeutic candidates that need to be efficiently evaluated early in the context of Phase I clinical trials. In this review, we discuss some of the key developments and novelties in the main histological groups.
Collapse
|
19
|
Patel JN. Lessons in practicing cancer genomics and precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1526081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Jai N. Patel
- Department of Cancer Pharmacology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| |
Collapse
|
20
|
Khan T, Stewart M, Blackman S, Rousseau R, Donoghue M, Cohen K, Seibel N, Fleury M, Benettaib B, Malik R, Vassal G, Reaman G. Accelerating Pediatric Cancer Drug Development: Challenges and Opportunities for Pediatric Master Protocols. Ther Innov Regul Sci 2018; 53:270-278. [PMID: 29759018 DOI: 10.1177/2168479018774533] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although outcomes for children with cancer have significantly improved over the past 40 years, there has been little progress in the treatment of some pediatric cancers, particularly when advanced. Additionally, clinical trial options and availability are often insufficient. Improved genomic and immunologic understanding of pediatric cancers, combined with innovative clinical trial designs, may provide an enhanced opportunity to study childhood cancers. Master protocols, which incorporate the use of precision medicine approaches, coupled with the ability to quickly assess the safety and effectiveness of new therapies, have the potential to accelerate early-phase clinical testing of novel therapeutics and which may result in more rapid approval of new drugs for children with cancer. Designing and conducting master protocols for children requires addressing similar principles and requirements as traditional adult oncology trials, but there are also unique considerations for master protocols conducted in children with cancer. The purpose of this paper is to define the key challenges and opportunities associated with this approach in order to ensure that master protocols can be adapted to benefit children and adolescents and ensure that adequate data are captured to advance, in parallel, the clinical development of investigational agents for children with cancer.
Collapse
Affiliation(s)
- Tahira Khan
- 1 Genentech Inc, a member of the Roche Group, South San Francisco, CA, USA
| | - Mark Stewart
- 2 Friends of Cancer Research, Washington, DC, USA
| | | | - Raphaël Rousseau
- 1 Genentech Inc, a member of the Roche Group, South San Francisco, CA, USA
| | | | - Kenneth Cohen
- 5 Department of Pediatrics and Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Nita Seibel
- 6 USA National Cancer Institute, Bethesda, MD, USA
| | - Mark Fleury
- 7 American Cancer Society Cancer Action Network Inc, Washington, DC, USA
| | | | | | - Gilles Vassal
- 10 Department of Clinical Research, Institut Gustave Roussy, Paris-Sud University, Paris, France
| | | |
Collapse
|
21
|
Venkatesan S, Hoogstraat M, Caljouw E, Pierson T, Spoor JKH, Zeneyedpour L, Dubbink HJ, Dekker LJ, van der Kaaij M, Kloezeman J, Berghauser Pont LME, Besselink NJM, Luider TM, Joore J, Martens JW, Lamfers MLM, Sleijfer S, Leenstra S. TP53 mutated glioblastoma stem-like cell cultures are sensitive to dual mTORC1/2 inhibition while resistance in TP53 wild type cultures can be overcome by combined inhibition of mTORC1/2 and Bcl-2. Oncotarget 2018; 7:58435-58444. [PMID: 27533080 PMCID: PMC5295441 DOI: 10.18632/oncotarget.11205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/26/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Glioblastoma is the most malignant tumor of the central nervous system and still lacks effective treatment. This study explores mutational biomarkers of 11 drugs targeting either the RTK/Ras/PI3K, the p53 or the Rb pathway using 25 patient-derived glioblastoma stem-like cell cultures (GSCs). RESULTS We found that TP53 mutated GSCs were approximately 3.5 fold more sensitive to dual inhibition of mammalian target of rapamycin complex 1 and 2 (mTORC1/2) compared to wild type GSCs. We identified that Bcl-2(Thr56/Ser70) phosphorylation contributed to the resistance of TP53 wild type GSCs against dual mTORC1/2 inhibition. The Bcl-2 inhibitor ABT-263 (navitoclax) increased sensitivity to the mTORC1/2 inhibitor AZD8055 in TP53 wild type GSCs, while sensitivity to AZD8055 in TP53 mutated GSCs remained unchanged. CONCLUSION Our data suggest that Bcl-2 confers resistance to mTORC1/2 inhibitors in TP53 wild type GSCs and that combined inhibition of both mTORC1/2 and Bcl-2 is worthwhile to explore further in TP53 wild type glioblastomas, whereas in TP53 mutated glioblastomas dual mTORC1/2 inhibitors should be explored.
Collapse
Affiliation(s)
| | - Marlous Hoogstraat
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Personalized Cancer Treatment (CPCT), University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Tessa Pierson
- Department of Neurosurgery, Brain Tumor Center Erasmus MC, Rotterdam, The Netherlands
| | - Jochem K H Spoor
- Department of Neurosurgery, Brain Tumor Center Erasmus MC, Rotterdam, The Netherlands
| | - Lona Zeneyedpour
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Hendrikus J Dubbink
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Lennard J Dekker
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Jenneke Kloezeman
- Department of Neurosurgery, Brain Tumor Center Erasmus MC, Rotterdam, The Netherlands
| | | | - Nicolle J M Besselink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Personalized Cancer Treatment (CPCT), University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Theo M Luider
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jos Joore
- Pepscope BV, Utrecht, The Netherlands
| | - John W Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.,Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Martine L M Lamfers
- Department of Neurosurgery, Brain Tumor Center Erasmus MC, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Center for Personalized Cancer Treatment (CPCT), University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.,Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center Erasmus MC, Rotterdam, The Netherlands.,Department of Neurosurgery, St. Elisabeth Hospital Tilburg, Tilburg, The Netherlands
| |
Collapse
|
22
|
Willis J, Vilar E. Pharmacogenomics: time to rethink its role in precision medicine. Ann Oncol 2018; 29:293-295. [DOI: 10.1093/annonc/mdx780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
23
|
Guo CC, Al-Ahmadie HA, Flaig TW, Kamat AM. Contribution of bladder cancer pathology assessment in planning clinical trials. Urol Oncol 2018; 39:713-719. [PMID: 29395955 DOI: 10.1016/j.urolonc.2018.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/30/2017] [Accepted: 01/03/2018] [Indexed: 11/25/2022]
Abstract
Bladder cancer is a heterogeneous disease that demonstrates a wide spectrum of histologic features. The modern classification of bladder cancer is largely based on pathologic analysis, which assesses tumor grade, stage, type, size, and other features that are essential for understanding the biological behavior of bladder cancer. Bladder cancers with similar histologic features are likely to show comparable responses to a new therapeutic agent in clinical trial. Furthermore, pathologic analysis also evaluates the quality of tissue samples in clinical trial to ensure the integrity of various molecular tests. In spite of the emerging role of genomic and molecular studies, pathology remains the cornerstone in the diagnosis, prognosis, and treatment of bladder cancer. Herein, the pathologic considerations for bladder cancer clinical trial planning are reviewed.
Collapse
Affiliation(s)
- Charles C Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Thomas W Flaig
- Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ashish M Kamat
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
24
|
Cardoso F, Harbeck N, Barrios CH, Bergh J, Cortés J, El Saghir N, Francis PA, Hudis CA, Ohno S, Partridge AH, Sledge GW, Smith IE, Gelmon KA. Research needs in breast cancer. Ann Oncol 2017; 28:208-217. [PMID: 27831505 DOI: 10.1093/annonc/mdw571] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
New research questions emerge as medical needs continue to evolve and as we improve our understanding of cancer biology and treatment of malignancies. Although significant advances have been made in some areas of breast cancer research resulting in improvements in therapies and outcomes over the last few decades, other areas have not benefited to the same degree and we continue to have many gaps in our knowledge. This article summarizes the 12 short and medium-term clinical research needs in breast cancer deemed as priorities in 2016 by a panel of experts, in an attempt to focus and accelerate future research in the most needed areas: (i) de-escalate breast cancer therapies in early breast cancer without sacrificing outcomes; (ii) explore optimal adjuvant treatment durations; (iii) develop better tools and strategies to identify patients with genetic predisposition; (iv) improve care in young patients with breast cancer; (v) develop tools to speed up drug development in biomarker-defined populations; (vi) identify and validate targets that mediate resistance to chemotherapy, endocrine therapy and anti-HER2 therapies; (vii) evaluate the efficacy of local-regional treatments for metastatic disease; (viii) better define the optimal sequence of treatments in the metastatic setting; (ix) evaluate the clinical impact of intra-patient heterogeneity (intra-tumor, inter-tumor and inter-lesion heterogeneity); (x) better understand the biology and identify new targets in triple-negative breast cancer; (xi) better understand immune surveillance in breast cancer and further develop immunotherapies; and (xii) increase survivorship research efforts including supportive care and quality of life.
Collapse
Affiliation(s)
- F Cardoso
- Breast Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | - N Harbeck
- Breast Center, Department of Obstetrics and Gynaecology, University of Munich (LMU), Munich, Germany
| | - C H Barrios
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - J Bergh
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - J Cortés
- Breast Cancer Unit, Ramon y Cajal University Hospital, Madrid.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - N El Saghir
- Department of Internal Medicine, NK Basile Cancer Institute American University of Beirut Medical Center, Beirut, Lebanon
| | - P A Francis
- Division of Cancer Medicine, Peter MacCallum Cancer Center, St Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - C A Hudis
- Chief Executive Officer, American Society of Clinical Oncology, Alexandria, USA
| | - S Ohno
- Center of Breast Oncology, Cancer Institute Hospital, Koto-Ku, Tokyo, Japan
| | - A H Partridge
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston
| | - G W Sledge
- Department of Medicine, Stanford University, Stanford, USA
| | - I E Smith
- Breast Unit, Royal Marsden Hospital, London, UK
| | - K A Gelmon
- Department of Medical Oncology, BC Cancer Agency, Vancouver, Canada
| |
Collapse
|
25
|
Manem VSK, Salgado R, Aftimos P, Sotiriou C, Haibe-Kains B. Network science in clinical trials: A patient-centered approach. Semin Cancer Biol 2017; 52:135-150. [PMID: 29278737 DOI: 10.1016/j.semcancer.2017.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 02/08/2023]
Abstract
There has been a paradigm shift in translational oncology with the advent of novel molecular diagnostic tools in the clinic. However, several challenges are associated with the integration of these sophisticated tools into clinical oncology and daily practice. High-throughput profiling at the DNA, RNA and protein levels (omics) generate a massive amount of data. The analysis and interpretation of these is non-trivial but will allow a more thorough understanding of cancer. Linear modelling of the data as it is often used today is likely to limit our understanding of cancer as a complex disease, and at times under-performs to capture a phenotype of interest. Network science and systems biology-based approaches, using machine learning and network science principles, that integrate multiple data sources, can uncover complex changes in a biological system. This approach will integrate a large number of potential biomarkers in preclinical studies to better inform therapeutic decisions and ultimately make substantial progress towards precision medicine. It will however require development of a new generation of clinical trials. Beyond discussing the challenges of high-throughput technologies, this review will develop a framework on how to implement a network science approach in new clinical trial designs in order to advance cancer care.
Collapse
Affiliation(s)
- Venkata S K Manem
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Brussels, Belgium; Department of Pathology, GZA Hospitals Antwerp, Belgium
| | - Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Brussels, Belgium; Medical Oncology Clinic, Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Haibe-Kains
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, Toronto, ON, Canada; Department of Computer Science, University of Toronto, Toronto, ON, Canada; Ontario Institute of Cancer Research, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
26
|
Tryfonidis K, Hartmann K, Morfouace M, Lacombe D. From bench to clinical trials the EORTC experience in biology-based clinical cancer research. J Egypt Natl Canc Inst 2017; 29:171-176. [PMID: 29122511 DOI: 10.1016/j.jnci.2017.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/06/2017] [Indexed: 10/18/2022] Open
Abstract
For over 50years the European Organization for Research and Treatment of Cancer (EORTC) has delivered major advances in cancer clinical research and cancer therapeutics. The introduction of molecularly targeted agents has led to significant improvements in outcome for patients with specific tumor types; however conventional chemotherapy remains the mainstay of treatment for the majority of patients. Due to increasing knowledge about the diversity of molecular pathways driving malignant progression, strategies to integrate biology into clinical research and development are continuously evolving. The challenges and the experience of the EORTC regarding how translational research is to be an indispensable component of the clinical research environment, which aims to deliver more sophisticated treatment approaches will be discussed in this perspective article.
Collapse
Affiliation(s)
- Konstantinos Tryfonidis
- European Organization for Research and Treatment of Cancer (EORTC), Av. Mounier 83/11, 1200 Brussels, Belgium.
| | - Katherine Hartmann
- European Organization for Research and Treatment of Cancer (EORTC), Av. Mounier 83/11, 1200 Brussels, Belgium
| | - Marie Morfouace
- European Organization for Research and Treatment of Cancer (EORTC), Av. Mounier 83/11, 1200 Brussels, Belgium
| | - Denis Lacombe
- European Organization for Research and Treatment of Cancer (EORTC), Av. Mounier 83/11, 1200 Brussels, Belgium
| |
Collapse
|
27
|
Sánchez NS, Mills GB, Mills Shaw KR. Precision oncology: neither a silver bullet nor a dream. Pharmacogenomics 2017; 18:1525-1539. [PMID: 29061079 DOI: 10.2217/pgs-2017-0094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Precision oncology is not an illusion, nor is it the magic bullet that will eradicate all cancers. Precision oncology is simply another weapon in our growing armament against cancer. Rather than honing in on the failures of a relatively young field, one should advocate for integrating its successes into widespread clinical practice, especially for indications, such as: ABL, ALK, BRAF, BRCA1, BRCA2, EGFR, KIT, KRAS, PDGFRA, PDGFRB, ROS1, BCR-ABL, FLT3 and ROS1, where aberrations have been shown to alter responses to US FDA approved drugs - that is, level 1 data. Moreover, to truly assess the promise of precision oncology, we must first begin by defining our expectations for this field. Importantly, we must recognize that the conception of precision oncology arose as an antithesis of the 'one-size fits all' cancer therapeutics approach. Consequently, tools used for evaluating these conventional, large-scale trials, are not directly transferable for assessing nonconventional, smaller-scale trials needed for evaluating precision oncology. Hence, a thorough vetting of precision oncology as another tool of the trade, must first begin by reassessing our expectations for this field, as well as current clinical trial designs and end point measurements. Importantly, we must recognize that most targeted therapy approaches are in their infancy, with only monotherapy approaches being assessed and combination therapies likely being necessary to fulfill the promise of precision oncology.
Collapse
Affiliation(s)
- Nora S Sánchez
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gordon B Mills
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenna R Mills Shaw
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
28
|
Allen CE, Laetsch TW, Mody R, Irwin MS, Lim MS, Adamson PC, Seibel NL, Parsons DW, Cho YJ, Janeway K. Target and Agent Prioritization for the Children's Oncology Group-National Cancer Institute Pediatric MATCH Trial. J Natl Cancer Inst 2017; 109:2972640. [PMID: 28376230 DOI: 10.1093/jnci/djw274] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/13/2016] [Indexed: 12/15/2022] Open
Abstract
Over the past decades, outcomes for children with cancer have improved dramatically through serial clinical trials based in large measure on dose intensification of cytotoxic chemotherapy for children with high-risk malignancies. Progress made through such dose intensification, in general, is no longer yielding further improvements in outcome. With the revolution in sequencing technologies and rapid development of drugs that block specific proteins and pathways, there is now an opportunity to improve outcomes for pediatric cancer patients through mutation-based targeted therapeutic strategies. The Children's Oncology Group (COG), in partnership with the National Cancer Institute (NCI), is planning a trial entitled the COG-NCI Pediatric Molecular Analysis for Therapeutic Choice (Pediatric MATCH) protocol utilizing an umbrella design. This protocol will have centralized infrastructure and will consist of a biomarker profiling protocol and multiple single-arm phase II trials of targeted therapies. Pediatric patients with recurrent or refractory solid tumors, lymphomas, or histiocytoses with measurable disease will be eligible. The Pediatric MATCH Target and Agent Prioritization (TAP) committee includes membership representing COG disease committees, the Food and Drug Administration, and the NCI. The TAP Committee systematically reviewed target and agent pairs for inclusion in the Pediatric MATCH trial. Fifteen drug-target pairs were reviewed by the TAP Committee, with seven recommended for further development as initial arms of the Pediatric MATCH trial. The current evidence for availability, efficacy, and safety of targeted agents in children for each class of mutation considered for inclusion in the Pediatric MATCH trial is discussed in this review.
Collapse
Affiliation(s)
- Carl E Allen
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Theodore W Laetsch
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Pauline Allen Gill Center for Cancer and Blood Disorders, Children's Health, Dallas, TX, USA
| | - Rajen Mody
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Meredith S Irwin
- Department of Pediatrics, Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Peter C Adamson
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nita L Seibel
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - D Williams Parsons
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Y Jae Cho
- Division of Pediatric Neurology, Doernbecher Children's Hospital, Portland, OR, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Katherine Janeway
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer Center and Blood Disorder Center, Boston, MA, USA
| | | |
Collapse
|
29
|
Liu J, Adhav R, Xu X. Current Progresses of Single Cell DNA Sequencing in Breast Cancer Research. Int J Biol Sci 2017; 13:949-960. [PMID: 28924377 PMCID: PMC5599901 DOI: 10.7150/ijbs.19627] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/08/2017] [Indexed: 12/16/2022] Open
Abstract
Breast cancers display striking genetic and phenotypic diversities. To date, several hypotheses are raised to explain and understand the heterogeneity, including theories for cancer stem cell (CSC) and clonal evolution. According to the CSC theory, the most tumorigenic cells, while maintaining themselves through symmetric division, divide asymmetrically to generate non-CSCs with less tumorigenic and metastatic potential, although they can also dedifferentiate back to CSCs. Clonal evolution theory recapitulates that a tumor initially arises from a single cell, which then undergoes clonal expansion to a population of cancer cells. During tumorigenesis and evolution process, cancer cells undergo different degrees of genetic instability and consequently obtain varied genetic aberrations. Yet the heterogeneity in breast cancers is very complex, poorly understood and subjected to further investigation. In recent years, single cell sequencing (SCS) technology developed rapidly, providing a powerful new way to better understand the heterogeneity, which may lay foundations to some new strategies for breast cancer therapies. In this review, we will summarize development of SCS technologies and recent advances of SCS in breast cancer.
Collapse
Affiliation(s)
- Jianlin Liu
- Faculty of Health Science, University of Macau, Macau SAR, China
| | - Ragini Adhav
- Faculty of Health Science, University of Macau, Macau SAR, China
| | - Xiaoling Xu
- Faculty of Health Science, University of Macau, Macau SAR, China
| |
Collapse
|
30
|
[Recovery of paraffin blocks and central archiving : Experiences of the Kiel lymph node registry and the German study group for Hodgkin lymphoma]. DER PATHOLOGE 2017; 38:529-534. [PMID: 28676898 DOI: 10.1007/s00292-017-0321-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Central collection of tissue blocks for pathological and translational research is particularly important in rare diseases. Transfer of tissue blocks from primary to central pathology is of crucial importance. OBJECTIVES We aimed to answer the following questions: Has the transfer of tissue blocks sent for consultation or within clinical trials changed over the last 20 years? What are the reasons for reclaiming tissue blocks by the primary pathology and what actions would convince primary pathologists to leave the blocks in the reference pathology? MATERIAL AND METHODS The first 100 biopsies of each year between 1995 and 2015 (n = 2100), as well as all tissue transfers within therapeutic studies (n = 1405, German Hodgkin Study Group, GHSG) between 1998 and 2015, were analyzed separately for block reclaims using the Department of Pathology database. A questionnaire evaluated the reasons for block reclaiming by the peripheral pathologists. RESULTS There is a significant increase in block reclaims during the period analyzed among submissions for consultation as well as in clinical trials (linear regression, p = 0.0195 and p = 0.0107). The percentage of block reclaims does not differ between consultations and cases submitted upon request within clinical trials (p = 0.2404, t-test). A survey among pathologies that reclaim the block showed that their willingness to leave the block at the reference center would increase if the compatibility with accreditation guidelines (39.3%), a positive statement from professional associations (25%), or a formal confirmation of availability (53.6%) is provided. DISCUSSION In particular, to improve research on rare diseases, it is desirable to point out the compatibility of central archiving in a designated center with accreditation guidelines.
Collapse
|
31
|
Horak P, Klink B, Heining C, Gröschel S, Hutter B, Fröhlich M, Uhrig S, Hübschmann D, Schlesner M, Eils R, Richter D, Pfütze K, Geörg C, Meißburger B, Wolf S, Schulz A, Penzel R, Herpel E, Kirchner M, Lier A, Endris V, Singer S, Schirmacher P, Weichert W, Stenzinger A, Schlenk RF, Schröck E, Brors B, von Kalle C, Glimm H, Fröhling S. Precision oncology based on omics data: The NCT Heidelberg experience. Int J Cancer 2017; 141:877-886. [PMID: 28597939 DOI: 10.1002/ijc.30828] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/24/2017] [Accepted: 05/29/2017] [Indexed: 12/17/2022]
Abstract
Precision oncology implies the ability to predict which patients will likely respond to specific cancer therapies based on increasingly accurate, high-resolution molecular diagnostics as well as the functional and mechanistic understanding of individual tumors. While molecular stratification of patients can be achieved through different means, a promising approach is next-generation sequencing of tumor DNA and RNA, which can reveal genomic alterations that have immediate clinical implications. Furthermore, certain genetic alterations are shared across multiple histologic entities, raising the fundamental question of whether tumors should be treated by molecular profile and not tissue of origin. We here describe MASTER (Molecularly Aided Stratification for Tumor Eradication Research), a clinically applicable platform for prospective, biology-driven stratification of younger adults with advanced-stage cancer across all histologies and patients with rare tumors. We illustrate how a standardized workflow for selection and consenting of patients, sample processing, whole-exome/genome and RNA sequencing, bioinformatic analysis, rigorous validation of potentially actionable findings, and data evaluation by a dedicated molecular tumor board enables categorization of patients into different intervention baskets and formulation of evidence-based recommendations for clinical management. Critical next steps will be to increase the number of patients that can be offered comprehensive molecular analysis through collaborations and partnering, to explore ways in which additional technologies can aid in patient stratification and individualization of treatment, to stimulate clinically guided exploratory research projects, and to gradually move away from assessing the therapeutic activity of targeted interventions on a case-by-case basis toward controlled clinical trials of genomics-guided treatments.
Collapse
Affiliation(s)
- Peter Horak
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Barbara Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Christoph Heining
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Gröschel
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Research Group Molecular Leukemogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Barbara Hutter
- Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | - Martina Fröhlich
- Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | - Sebastian Uhrig
- Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | - Daniel Hübschmann
- Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany.,Department of Pediatric Immunology, Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Roland Eils
- Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany.,Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology and BioQuant, Heidelberg University, Heidelberg, Germany
| | - Daniela Richter
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katrin Pfütze
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-Heidelberg Center for Personalized Oncology (HIPO), Heidelberg, Germany
| | - Christina Geörg
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-Heidelberg Center for Personalized Oncology (HIPO), Heidelberg, Germany
| | - Bettina Meißburger
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-Heidelberg Center for Personalized Oncology (HIPO), Heidelberg, Germany
| | - Stephan Wolf
- Genomics and Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Angela Schulz
- Genomics and Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Roland Penzel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martina Kirchner
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Amelie Lier
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Volker Endris
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephan Singer
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Schirmacher
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technische Universität München, Munich, Germany.,DKTK, Munich, Germany
| | - Albrecht Stenzinger
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Evelin Schröck
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Benedikt Brors
- DKTK, Heidelberg, Germany.,Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany.,DKTK, Heidelberg, Germany.,DKFZ-Heidelberg Center for Personalized Oncology (HIPO), Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| |
Collapse
|
32
|
Abstract
INTRODUCTION Nasopharyngeal carcinoma (NPC) is endemic to Southern China and Asia and is etiologically associated with the Epstein Barr virus (EBV). Whole exome and genome sequencing (WES, WGS) studies of NPC have reported several actionable therapeutic targets, and that the mutational load of NPC maybe comparable to that of squamous head and neck cancer. These unique biological characteristics have been exploited as potential targets and a wide range of investigational drugs are being investigated in clinical trials. Area covered: This review focused on the latest clinical development of the most promising classes of investigational agents in the treatment of advanced NPC. These include inhibitors of tumor angiogenesis, kinase signaling pathways and immunotherapy. Expert opinion: Checkpoint inhibitors and EBV-specific T-cell therapy have shown promising activity in early phase clinical trials, and are being further evaluated in randomized studies. For patients whose tumors express genetic alterations that are known to predict response to kinase inhibitors, novel trial designs such as an 'Umbrella' study may be considered given the abundance of targeted agents that are now available for clinical evaluation. It is envisioned that regulatory approval for new drugs for advanced NPC will occur in the near future.
Collapse
Affiliation(s)
- Brigette B Y Ma
- a State Key Laboratory in Oncology in South China , Sir YK Pao Centre for Cancer , Hong Kong SAR , China.,b Department of Clinical Oncology , Hong Kong Cancer Institute , Hong Kong SAR , China.,c Li Ka Shing Institute of Health Sciences , The Chinese University of Hong Kong , Hong Kong SAR , China
| | - Edwin P Hui
- a State Key Laboratory in Oncology in South China , Sir YK Pao Centre for Cancer , Hong Kong SAR , China.,b Department of Clinical Oncology , Hong Kong Cancer Institute , Hong Kong SAR , China.,c Li Ka Shing Institute of Health Sciences , The Chinese University of Hong Kong , Hong Kong SAR , China
| | - Anthony T C Chan
- a State Key Laboratory in Oncology in South China , Sir YK Pao Centre for Cancer , Hong Kong SAR , China.,b Department of Clinical Oncology , Hong Kong Cancer Institute , Hong Kong SAR , China.,c Li Ka Shing Institute of Health Sciences , The Chinese University of Hong Kong , Hong Kong SAR , China
| |
Collapse
|
33
|
Zick A, Peretz T, Lotem M, Hubert A, Katz D, Temper M, Rottenberg Y, Uziely B, Nechushtan H, Meirovitz A, Sonnenblick A, Sapir E, Edelman D, Goldberg Y, Lossos A, Rosenberg S, Fried I, Finklstein R, Pikarsky E, Goldshmidt H. Treatment inferred from mutations identified using massive parallel sequencing leads to clinical benefit in some heavily pretreated cancer patients. Medicine (Baltimore) 2017; 96:e6931. [PMID: 28514312 PMCID: PMC5440149 DOI: 10.1097/md.0000000000006931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Molecular portraits of numerous tumors have flooded oncologists with vast amounts of data. In parallel, effective inhibitors of central pathways have shown great clinical benefit. Together, this promises potential clinical benefits to otherwise end-stage cancer patients. Here, we report a clinical service offering mutation detection of archived samples using the ion Ampliseq cancer panel coupled with clinical consultation.A multidisciplinary think tank consisting of oncologists, molecular-biologists, genetic counselors, and pathologists discussed 67 heavily pretreated, advanced cancer patient cases, taking into account mutations identified using ion Ampliseq cancer panel, medical history, and relevant literature.The team generated a treatment plan, targeting specific mutations, for 41 out of 64 cases. Three patients died before results were available. For 32 patients, the treating oncologists chose not to include the panel recommendation in the treatment plan for various reasons. Nine patients were treated as recommended by the panel, 5 with clinical benefit, and 4 with disease progression.This study suggests that routine use of massive parallel tumor sequencing is feasible and can judiciously affect treatment decisions when coupled with multidisciplinary team-based decision making. Administration of personalized based therapies at an earlier stage of disease, expansion of genetic alterations examined, and increased availability of targeted therapies may lead to further improvement in the clinical outcome of metastatic cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Alexander Lossos
- Leslie and Michael Gaffin Center for Oncology, Departments of Oncology and Neurology, Hebrew University-Hadassah Medical Center
| | - Shai Rosenberg
- Leslie and Michael Gaffin Center for Oncology, Departments of Oncology and Neurology, Hebrew University-Hadassah Medical Center
| | - Iris Fried
- Department of Pediatrics, Hadassah Medical Center, Division of Pediatric Hematology and Oncology
| | - Ruth Finklstein
- Lautenberg Center for Immunology, IMRIC, The Hebrew University-Hadassah Medical School
| | - Eli Pikarsky
- Lautenberg Center for Immunology, IMRIC, The Hebrew University-Hadassah Medical School
- Department of Pathology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Hanoch Goldshmidt
- Department of Pathology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
34
|
Tian H, Zhou C, Yang J, Li J, Gong Z. Long and short noncoding RNAs in lung cancer precision medicine: Opportunities and challenges. Tumour Biol 2017; 39:1010428317697578. [PMID: 28381159 DOI: 10.1177/1010428317697578] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The long and short noncoding RNAs have been involved in the molecular diagnosis, targeted therapy, and predicting prognosis of lung cancer. Utilizing noncoding RNAs as biomarkers and systemic RNA interference as an innovative therapeutic strategy has an immense likelihood to generate novel concepts in precision oncology. Targeting of RNA interference payloads such as small interfering RNAs, microRNA mimetic, or anti-microRNA (antagomirs) into specific cell types has achieved initial success. The clinical trials of noncoding RNA-based therapies are on the way with some positive results. Many attempts are done for developing novel noncoding RNA delivery strategies that could overcome systemic or local barriers. Furthermore, it precipitates concerted efforts to define the molecular subtypes of lung cancer, characterize the genomic landscape of lung cancer subtypes, identify novel therapeutic targets, and reveal mechanisms of sensitivity and resistance to targeted therapies. These efforts contribute a visible effect now in lung cancer precision medicine: patients receive molecular testing to determine whether their tumor harbors an actionable come resistance to the first-generation drugs are in clinical trials, and drugs targeting the immune system are showing activity in patients. This extraordinary promise is tempered by the sobering fact that even the newest treatments for metastatic disease are rarely curative and are effective only in a small fraction of all patients. Thus, ongoing and future efforts to find new vulnerabilities of lung cancers unravel the complexity of drug resistance, increase the efficacy of immunotherapies, and perform biomarker-driven clinical trials are necessary to improve the outcome of lung cancer patients.
Collapse
Affiliation(s)
- Haihua Tian
- 1 Department of Biochemistry and Molecular Biology, Ningbo University School of Medicine, Ningbo, China.,2 Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China.,3 Department of Laboratory Medicine, Ningbo Kangning Hospital, Ningbo, China
| | - Chengwei Zhou
- 4 Department of Thoracic Surgery, The Affiliated Hospital of Ningbo University School of Medicine, Ningbo, China
| | - Jie Yang
- 1 Department of Biochemistry and Molecular Biology, Ningbo University School of Medicine, Ningbo, China.,2 Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
| | - Jingqiu Li
- 1 Department of Biochemistry and Molecular Biology, Ningbo University School of Medicine, Ningbo, China.,2 Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
| | - Zhaohui Gong
- 1 Department of Biochemistry and Molecular Biology, Ningbo University School of Medicine, Ningbo, China.,2 Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
| |
Collapse
|
35
|
Hsieh JJ, Manley BJ, Khan N, Gao J, Carlo MI, Cheng EH. Overcome tumor heterogeneity-imposed therapeutic barriers through convergent genomic biomarker discovery: A braided cancer river model of kidney cancer. Semin Cell Dev Biol 2017; 64:98-106. [PMID: 27615548 PMCID: PMC5522717 DOI: 10.1016/j.semcdb.2016.09.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022]
Abstract
Tumor heterogeneity, encompassing genetic, epigenetic, and microenvironmental variables, is extremely complex and presents challenges to cancer diagnosis and therapy. Genomic efforts on genetic intratumor heterogeneity (G-ITH) confirm branched evolution, support the trunk-branch cancer model, and present a seemingly insurmountable obstacle to conquering cancers. G-ITH is conspicuous in clear cell renal cell carcinoma (ccRCC), where its presence complicates identification and validation of biomarkers and thwarts efforts in advancing precision cancer therapeutics. However, long-term clinical benefits on targeted therapy are not uncommon in metastatic ccRCC patients, implicating that there are underlying constraints during ccRCC evolution, which in turn force a nonrandom sequence of parallel gene/pathway/function/phenotype convergence within individual tumors. Accordingly, we proposed a "braided cancer river model" depicting ccRCC evolution, which deduces cancer development based on multiregion tumor genomics of exceptional mTOR inhibitor (mTORi) responders. Furthermore, we employ an outlier case to explore the river model and highlight the importance of "Five NGS Matters: Number, Frequency, Position, Site and Time" in assessing cancer genomics for precision medicine. This mutable cancer river model may capture clinically significant phenotype-convergent events, predict vulnerability/resistance mechanisms, and guide effective therapeutic strategies. Our model originates from studying exceptional responders in ccRCC, which warrants further refinement and future validation concerning its applicability to other cancer types. The goal of this review is employing kidney cancer as an example to illustrate critical issues concerning tumor heterogeneity.
Collapse
Affiliation(s)
- James J Hsieh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Brandon J Manley
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Nabeela Khan
- Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY11203, United States
| | - JianJiong Gao
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| |
Collapse
|
36
|
Li B, Eschrich SA, Berglund A, Mitchell M, Fenstermacher D, Danaee H, Dai H, Sullivan D, Trepicchio WL, Dalton WS. Use of the Total Cancer Care System to Enrich Screening for CD30-Positive Solid Tumors for Patient Enrollment Into a Brentuximab Vedotin Clinical Trial: A Pilot Study to Evaluate Feasibility. JMIR Res Protoc 2017; 6:e45. [PMID: 28320689 PMCID: PMC5379017 DOI: 10.2196/resprot.7289] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/22/2017] [Accepted: 02/22/2017] [Indexed: 11/13/2022] Open
Abstract
Background One approach to identify patients who meet specific eligibility criteria for target-based clinical trials is to use patient and tumor registries to prescreen patient populations. Objective Here we demonstrate that the Total Cancer Care (TCC) Protocol, an ongoing, observational study, may provide a solution for rapidly identifying patients with CD30-positive tumors eligible for CD30-targeted therapies such as brentuximab vedotin. Methods The TCC patient gene expression profiling database was retrospectively screened for CD30 gene expression determined using HuRSTA-2a520709 Affymetrix arrays (GPL15048). Banked tumor tissue samples were used to determine CD30 protein expression by semiquantitative immunohistochemistry. Statistical comparisons of Z- and H-scores were performed using R statistical software (The R Foundation), and the predictive value, accuracy, sensitivity, and specificity of CD30 gene expression versus protein expression was estimated. Results As of March 2015, 120,887 patients have consented to the institutional review board–approved TCC Protocol. A total of 39,157 fresh frozen tumor specimens have been collected, from which over 14,000 samples have gene expression data available. CD30 RNA was expressed in a number of solid tumors; the highest median CD30 RNA expression was observed in primary tumors from lymph node, soft tissue (many sarcomas), lung, skin, and esophagus (median Z-scores 1.011, 0.399, 0.202, 0.152, and 1.011, respectively). High level CD30 gene expression significantly enriches for CD30-positive protein expression in breast, lung, skin, and ovarian cancer; accuracy ranged from 72% to 79%, sensitivity from 75% to 100%, specificity from 70% to 76%, positive predictive value from 20% to 40%, and negative predictive value from 95% to 100%. Conclusions The TCC gene expression profiling database guided tissue selection that enriched for CD30 protein expression in a number of solid tumor types. Such an approach may improve screening efficiency for enrolling patients into biomarker-based clinical trials.
Collapse
Affiliation(s)
- Bin Li
- Takeda Pharmaceuticals International Company, Takeda Data Science Institute, Cambridge, MA, United States
| | - Steven A Eschrich
- H Lee Moffitt Cancer Center and Research Institute, Biostatistics and Bioinformatics, Tampa, FL, United States
| | - Anders Berglund
- H Lee Moffitt Cancer Center and Research Institute, Biostatistics and Bioinformatics, Tampa, FL, United States
| | | | | | - Hadi Danaee
- Takeda Pharmaceuticals International Company, Translational and Biomarker Research, Cambridge, MA, United States
| | - Hongyue Dai
- M2Gen, Bioinformatics, Tampa, FL, United States
| | - Daniel Sullivan
- H Lee Moffitt Cancer Center and Research Institute, Blood and Marrow Transplantation, Tampa, FL, United States
| | - William L Trepicchio
- Takeda Pharmaceuticals International Company, Translational and Biomarker Research, Cambridge, MA, United States
| | | |
Collapse
|
37
|
Li W, Chen C, Li X, Beckman RA. Estimation of treatment effect in two-stage confirmatory oncology trials of personalized medicines. Stat Med 2017; 36:1843-1861. [PMID: 28303586 DOI: 10.1002/sim.7272] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/14/2017] [Indexed: 12/26/2022]
Abstract
A personalized medicine may benefit a subpopulation with certain predictive biomarker signatures or certain disease types. However, there is great uncertainty about drug activity in a subpopulation when designing a confirmatory trial in practice, and it is logical to take a two-stage approach with the study unless credible external information is available for decision-making purpose. The first stage deselects (or prunes) non-performing subpopulations at an interim analysis, and the second stage pools the remaining subpopulations in the final analysis. The endpoints used at the two stages can be different in general. A key issue of interest is the statistical property of the test statistics and point estimate at the final analysis. Previous research has focused on type I error control and power calculation for such two-stage designs. This manuscript will investigate estimation bias of the treatment effect, which is implicit in the adjustment of nominal type I error for multiplicity control in such two-stage designs. Previous work handles the treatment effect of an intermediate endpoint as a nuisance parameter to provide the most conservative type I error control. This manuscript takes the same approach to explore the bias. The methodology is applied to the two previously studied designs. In the first design, patients with different biomarker levels are enrolled in a study, and the treatment effect is assumed to be in an order. The goal of the interim analysis is to identify a biomarker cut-off point for the subpopulations. In the second design, patients with different tumour types but the same biomarker signature are included in a trial applying a basket design. The goal of the interim analysis is to identify a subset of tumour types in the absence of treatment effect ordering. Closed-form equations are provided for the estimation bias as well as the variance under the two designs. Simulations are conducted under various scenarios to validate the analytic results that demonstrated that the bias can be properly estimated in practice. Worked examples are presented. Extensions to general adaptive designs and operational considerations are discussed. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Wen Li
- Biostatistics and Research Decision Sciences, Merck Research Laboratories (MRL), Merck & Co., Inc, Kenilworth, NJ, U.S.A
| | - Cong Chen
- Biostatistics and Research Decision Sciences, Merck Research Laboratories (MRL), Merck & Co., Inc, Kenilworth, NJ, U.S.A
| | - Xiaoyun Li
- Biostatistics and Research Decision Sciences, Merck Research Laboratories (MRL), Merck & Co., Inc, Kenilworth, NJ, U.S.A
| | - Robert A Beckman
- Departments of Oncology and of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, 2115 Wisconsin Avenue, Suite 110, Washington, DC, 20007, U.S.A
| |
Collapse
|
38
|
Saad ED, Paoletti X, Burzykowski T, Buyse M. Precision medicine needs randomized clinical trials. Nat Rev Clin Oncol 2017; 14:317-323. [DOI: 10.1038/nrclinonc.2017.8] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
39
|
Tissue-based next generation sequencing: application in a universal healthcare system. Br J Cancer 2017; 116:553-560. [PMID: 28103613 PMCID: PMC5344287 DOI: 10.1038/bjc.2016.452] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/17/2016] [Accepted: 11/30/2016] [Indexed: 02/07/2023] Open
Abstract
In the context of solid tumours, the evolution of cancer therapies to more targeted and nuanced approaches has led to the impetus for personalised medicine. The targets for these therapies are largely based on the driving genetic mutations of the tumours. To track these multiple driving mutations the use of next generation sequencing (NGS) coupled with a morphomolecular approach to tumours, has the potential to deliver on the promises of personalised medicine. A review of NGS and its application in a universal healthcare (UHC) setting is undertaken as the technology has a wide appeal and utility in diagnostic, clinical trial and research paradigms. Furthermore, we suggest that these can be accommodated with a unified integromic approach. Challenges remain in bringing NGS to routine clinical use and these include validation, handling of the large amounts of information flow and production of a clinically useful report. These challenges are particularly acute in the setting of UHC where tests are not reimbursed and there are finite resources available. It is our opinion that the challenges faced in applying NGS in a UHC setting are surmountable and we outline our approach for its routine application in diagnostic, clinical trial and research paradigms.
Collapse
|
40
|
Matrisian LM, Berlin JD. The Past, Present, and Future of Pancreatic Cancer Clinical Trials. Am Soc Clin Oncol Educ Book 2017; 35:e205-15. [PMID: 27249725 DOI: 10.1200/edbk_159117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Upper gastrointestinal malignancies comprise half of the deadliest cancers as defined by those with a 5-year survival rate less than 50%. Using pancreatic adenocarcinoma (PAC) as an example, we retrospectively evaluated the success of phase III clinical trials, examined the current landscape of clinical trials, and identified emerging areas that foretell the future for this disease. Pancreatic and liver cancers are on the rise and will be the second and third leading causes of cancer deaths in 2030. A total of 35 different agents or combinations have been tested in randomized phase III clinical trials for patients with advanced PAC over the past 25 years, but only 11% have been incorporated into clinical practice. There has been a 37% increase in the number of PAC trials open in the United States between 2011 to 2012 and 2014 to 2015. Enrollment has also increased slightly, from 3.85% of the newly diagnosed cases in 2011 to 4.15% in 2014. However, the demand for patients far exceeds the number of patients available for these trials. On the horizon is the realization that stratification of patients with PAC using biomarkers that predict a high probability of a response could reallocate patients to faster, smaller trials with a greater chance of a survival benefit. The current landscape of PAC clinical trials and the launch of the Pancreatic Cancer Action Network's Know Your Tumor initiative indicate this shift is starting to occur, with particular emphasis on targeted therapies, immunotherapies, and agents that disrupt the stroma.
Collapse
Affiliation(s)
- Lynn M Matrisian
- From the Pancreatic Cancer Action Network, Manhattan Beach, CA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN
| | - Jordan D Berlin
- From the Pancreatic Cancer Action Network, Manhattan Beach, CA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN
| |
Collapse
|
41
|
Renfro LA, Sargent DJ. Statistical controversies in clinical research: basket trials, umbrella trials, and other master protocols: a review and examples. Ann Oncol 2017; 28:34-43. [PMID: 28177494 PMCID: PMC5834138 DOI: 10.1093/annonc/mdw413] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In recent years, cancers once viewed as relatively homogeneous in terms of organ location and treatment strategy are now better understood to be increasingly heterogeneous across biomarker and genetically defined patient subgroups. This has produced a shift toward development of biomarker-targeted agents during a time when funding for cancer research has been limited; as a result, the need for improved operational efficiency in studying many agent-and-target combinations in parallel has emerged. Platform trials, basket trials, and umbrella trials are new approaches to clinical research driven by this need for enhanced efficiency in the modern era of increasingly specific cancer subpopulations and decreased resources to study treatments for individual cancer subtypes in a traditional way. In this review, we provide an overview of these new types of clinical trial designs, including discussions of motivation for their use, recommended terminology, examples, and challenges encountered in their application.
Collapse
Affiliation(s)
- L. A. Renfro
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, USA
| | - D. J. Sargent
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, USA
| |
Collapse
|
42
|
Borad MJ, Egan JB, Condjella RM, Liang WS, Fonseca R, Ritacca NR, McCullough AE, Barrett MT, Hunt KS, Champion MD, Patel MD, Young SW, Silva AC, Ho TH, Halfdanarson TR, McWilliams RR, Lazaridis KN, Ramanathan RK, Baker A, Aldrich J, Kurdoglu A, Izatt T, Christoforides A, Cherni I, Nasser S, Reiman R, Cuyugan L, McDonald J, Adkins J, Mastrian SD, Valdez R, Jaroszewski DE, Von Hoff DD, Craig DW, Stewart AK, Carpten JD, Bryce AH. Clinical Implementation of Integrated Genomic Profiling in Patients with Advanced Cancers. Sci Rep 2016; 6:25. [PMID: 28003660 PMCID: PMC5431338 DOI: 10.1038/s41598-016-0021-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/02/2016] [Indexed: 12/20/2022] Open
Abstract
DNA focused panel sequencing has been rapidly adopted to assess therapeutic targets in advanced/refractory cancer. Integrated Genomic Profiling (IGP) utilising DNA/RNA with tumour/normal comparisons in a Clinical Laboratory Improvement Amendments (CLIA) compliant setting enables a single assay to provide: therapeutic target prioritisation, novel target discovery/application and comprehensive germline assessment. A prospective study in 35 advanced/refractory cancer patients was conducted using CLIA-compliant IGP. Feasibility was assessed by estimating time to results (TTR), prioritising/assigning putative therapeutic targets, assessing drug access, ascertaining germline alterations, and assessing patient preferences/perspectives on data use/reporting. Therapeutic targets were identified using biointelligence/pathway analyses and interpreted by a Genomic Tumour Board. Seventy-five percent of cases harboured 1–3 therapeutically targetable mutations/case (median 79 mutations of potential functional significance/case). Median time to CLIA-validated results was 116 days with CLIA-validation of targets achieved in 21/22 patients. IGP directed treatment was instituted in 13 patients utilising on/off label FDA approved drugs (n = 9), clinical trials (n = 3) and single patient IND (n = 1). Preliminary clinical efficacy was noted in five patients (two partial response, three stable disease). Although barriers to broader application exist, including the need for wider availability of therapies, IGP in a CLIA-framework is feasible and valuable in selection/prioritisation of anti-cancer therapeutic targets.
Collapse
Affiliation(s)
- Mitesh J Borad
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA. .,Mayo Clinic Cancer Center, Scottsdale, AZ, USA. .,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Jan B Egan
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Winnie S Liang
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Rafael Fonseca
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA.,Mayo Clinic Cancer Center, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Michael T Barrett
- Mayo Clinic Cancer Center, Scottsdale, AZ, USA.,Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Katherine S Hunt
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA
| | - Mia D Champion
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Biomedical Statistics and Informatics, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Scott W Young
- Department of Radiology, Mayo Clinic, Scottsdale, AZ, USA
| | - Alvin C Silva
- Department of Radiology, Mayo Clinic, Scottsdale, AZ, USA
| | - Thai H Ho
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA.,Mayo Clinic Cancer Center, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thorvardur R Halfdanarson
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA.,Mayo Clinic Cancer Center, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Robert R McWilliams
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Cancer Center, Rochester, MN, USA
| | | | - Ramesh K Ramanathan
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA.,Mayo Clinic Cancer Center, Scottsdale, AZ, USA
| | - Angela Baker
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | - Ahmet Kurdoglu
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Tyler Izatt
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | - Irene Cherni
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Sara Nasser
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Rebecca Reiman
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Lori Cuyugan
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | | | | | | | | | | - David W Craig
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - A Keith Stewart
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA.,Mayo Clinic Cancer Center, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - John D Carpten
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Alan H Bryce
- Division of Hematology/Oncology Mayo Clinic, Scottsdale, AZ, USA.,Mayo Clinic Cancer Center, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
43
|
Horak P, Fröhling S, Glimm H. Integrating next-generation sequencing into clinical oncology: strategies, promises and pitfalls. ESMO Open 2016; 1:e000094. [PMID: 27933214 PMCID: PMC5133384 DOI: 10.1136/esmoopen-2016-000094] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 12/24/2022] Open
Abstract
We live in an era of genomic medicine. The past five years brought about many significant achievements in the field of cancer genetics, driven by rapidly evolving technologies and plummeting costs of next-generation sequencing (NGS). The official completion of the Cancer Genome Project in 2014 led many to envision the clinical implementation of cancer genomic data as the next logical step in cancer therapy. Stemming from this vision, the term 'precision oncology' was coined to illustrate the novelty of this individualised approach. The basic assumption of precision oncology is that molecular markers detected by NGS will predict response to targeted therapies independently from tumour histology. However, along with a ubiquitous availability of NGS, the complexity and heterogeneity at the individual patient level had to be acknowledged. Not only does the latter present challenges to clinical decision-making based on sequencing data, it is also an obstacle to the rational design of clinical trials. Novel tissue-agnostic trial designs were quickly developed to overcome these challenges. Results from some of these trials have recently demonstrated the feasibility and efficacy of this approach. On the other hand, there is an increasing amount of whole-exome and whole-genome NGS data which allows us to assess ever smaller differences between individual patients with cancer. In this review, we highlight different tumour sequencing strategies currently used for precision oncology, describe their individual strengths and weaknesses, and emphasise their feasibility in different clinical settings. Further, we evaluate the possibility of NGS implementation in current and future clinical trials, and point to the significance of NGS for translational research.
Collapse
Affiliation(s)
- Peter Horak
- Department of Translational Oncology , National Center for Tumor Diseases Heidelberg, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Stefan Fröhling
- Department of Translational Oncology , National Center for Tumor Diseases Heidelberg, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Hanno Glimm
- Department of Translational Oncology , National Center for Tumor Diseases Heidelberg, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
44
|
Stockley TL, Oza AM, Berman HK, Leighl NB, Knox JJ, Shepherd FA, Chen EX, Krzyzanowska MK, Dhani N, Joshua AM, Tsao MS, Serra S, Clarke B, Roehrl MH, Zhang T, Sukhai MA, Califaretti N, Trinkaus M, Shaw P, van der Kwast T, Wang L, Virtanen C, Kim RH, Razak ARA, Hansen AR, Yu C, Pugh TJ, Kamel-Reid S, Siu LL, Bedard PL. Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial. Genome Med 2016; 8:109. [PMID: 27782854 PMCID: PMC5078968 DOI: 10.1186/s13073-016-0364-2] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 10/11/2016] [Indexed: 12/23/2022] Open
Abstract
Background The clinical utility of molecular profiling of tumor tissue to guide treatment of patients with advanced solid tumors is unknown. Our objectives were to evaluate the frequency of genomic alterations, clinical “actionability” of somatic variants, enrollment in mutation-targeted or other clinical trials, and outcome of molecular profiling for advanced solid tumor patients at the Princess Margaret Cancer Centre (PM). Methods Patients with advanced solid tumors aged ≥18 years, good performance status, and archival tumor tissue available were prospectively consented. DNA from archival formalin-fixed paraffin-embedded tumor tissue was tested using a MALDI-TOF MS hotspot panel or a targeted next generation sequencing (NGS) panel. Somatic variants were classified according to clinical actionability and an annotated report included in the electronic medical record. Oncologists were provided with summary tables of their patients’ molecular profiling results and available mutation-specific clinical trials. Enrolment in genotype-matched versus genotype-unmatched clinical trials following release of profiling results and response by RECIST v1.1 criteria were evaluated. Results From March 2012 to July 2014, 1893 patients were enrolled and 1640 tested. After a median follow-up of 18 months, 245 patients (15 %) who were tested were subsequently treated on 277 therapeutic clinical trials, including 84 patients (5 %) on 89 genotype-matched trials. The overall response rate was higher in patients treated on genotype-matched trials (19 %) compared with genotype-unmatched trials (9 %; p < 0.026). In a multi-variable model, trial matching by genotype (p = 0.021) and female gender (p = 0.034) were the only factors associated with increased likelihood of treatment response. Conclusions Few advanced solid tumor patients enrolled in a prospective institutional molecular profiling trial were treated subsequently on genotype-matched therapeutic trials. In this non-randomized comparison, genotype-enrichment of early phase clinical trials was associated with an increased objective tumor response rate. Trial registration NCT01505400 (date of registration 4 January 2012). Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0364-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tracy L Stockley
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Amit M Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Hal K Berman
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Natasha B Leighl
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Jennifer J Knox
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Frances A Shepherd
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Eric X Chen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Monika K Krzyzanowska
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Neesha Dhani
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Anthony M Joshua
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Ming-Sound Tsao
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Stefano Serra
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Blaise Clarke
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Michael H Roehrl
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Tong Zhang
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| | - Mahadeo A Sukhai
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| | - Nadia Califaretti
- Department of Oncology, Grand River Regional Cancer Centre, Kitchener-Waterloo, Canada.,Department of Oncology, McMaster University, Faculty of Health Sciences, Hamilton, Canada
| | - Mateya Trinkaus
- Department of Medicine, Markham Stouffville Hospital, Markham, Canada
| | - Patricia Shaw
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Theodorus van der Kwast
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Lisa Wang
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, Canada
| | - Carl Virtanen
- Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada.,Princess Margaret Research Institute, Princess Margaret Cancer Centre, Toronto, Canada
| | - Raymond H Kim
- Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada.,Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Albiruni R A Razak
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Aaron R Hansen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Celeste Yu
- Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Trevor J Pugh
- Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Princess Margaret Research Institute, Princess Margaret Cancer Centre, Toronto, Canada
| | - Suzanne Kamel-Reid
- Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Lillian L Siu
- Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada.,Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Philippe L Bedard
- Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, Canada. .,Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, M5G 2M9, Canada. .,Department of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
45
|
Beckman RA, Antonijevic Z, Kalamegham R, Chen C. Adaptive Design for a Confirmatory Basket Trial in Multiple Tumor Types Based on a Putative Predictive Biomarker. Clin Pharmacol Ther 2016; 100:617-625. [PMID: 27509351 DOI: 10.1002/cpt.446] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 07/28/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Increasingly, tumors are defined on a molecular basis rather than only on histology, and targeted agents, which address these molecular subtypes, are being approved. This profusion of molecular subtypes creates "rare" diseases as subsets of common cancers, leading to difficulties in enrolling sufficiently large cohorts for confirmatory trials. However, if the molecular subtype is shared across various histologies, these may be pooled into a basket trial. To date, basket trials have been primarily for exploratory early development. In this perspective, we consider qualitative designs for confirmatory basket trials. These confirmatory basket designs will provide patients in niche indications with enhanced access to novel therapies, facilitate development and full approval for niche indications, allow accelerated approval for indications within a basket based on a surrogate endpoint, reduce development cost by combining trials, and enhance the ability of regulatory authorities to evaluate risk and benefit in niche indications.
Collapse
Affiliation(s)
- R A Beckman
- Departments of Oncology and of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
| | | | - R Kalamegham
- American Association for Cancer Research, Office of Science Policy and Government Affairs, Washington, DC, USA.,Current address: Genentech, Washington, DC, USA
| | - C Chen
- Biostatistics and Research Decision Sciences, Merck Research Laboratories, Kenilworth, New Jersey, USA
| |
Collapse
|
46
|
Chen C, Li X(N, Yuan S, Antonijevic Z, Kalamegham R, Beckman RA. Statistical Design and Considerations of a Phase 3 Basket Trial for Simultaneous Investigation of Multiple Tumor Types in One Study. Stat Biopharm Res 2016. [DOI: 10.1080/19466315.2016.1193044] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Cong Chen
- Biostatistics and Research Decision Sciences, Merck Research Laboratories, Upper Gwynedd, PA, USA
| | - Xiaoyun (Nicole) Li
- Biostatistics and Research Decision Sciences, Merck Research Laboratories, Upper Gwynedd, PA, USA
| | - Shuai Yuan
- Biostatistics and Research Decision Sciences, Merck Research Laboratories, Upper Gwynedd, PA, USA
| | | | - Rasika Kalamegham
- American Association for Cancer Research, Office of Science Policy and Government Affairs, Washington, DC, USA
| | - Robert A. Beckman
- Departments of Oncology and of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
47
|
Rodriguez-Rodriguez L, Hirshfield KM, Rojas V, DiPaola RS, Gibbon D, Hellmann M, Isani S, Leiser A, Riedlinger GM, Wagreich A, Ali SM, Elvin JA, Miller VA, Ganesan S. Use of comprehensive genomic profiling to direct point-of-care management of patients with gynecologic cancers. Gynecol Oncol 2016; 141:2-9. [PMID: 27016222 DOI: 10.1016/j.ygyno.2016.02.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/18/2016] [Accepted: 02/21/2016] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To determine the feasibility and clinical utility of using comprehensive genomic profiling (CGP) in the course of clinical care to identify clinically relevant tumor genomic alterations for patients with either rare or refractory gynecologic cancers to facilitate point-of-care management. Use of an expert, multidisciplinary, institutional molecular tumor board (MTB) assessment is discussed regarding input on putative targeted options for individualized therapy. METHODS A prospective clinical trial is ongoing. We report on the initial 69 patients with gynecologic cancers that were either rare or refractory to standard therapy. CGP was performed by Foundation Medicine, Inc. Genomic alterations were reviewed by members of an MTB. Consensus recommendations on genomically targeted, FDA-approved, on- and off-label therapies and clinical trials were sent to the treating physician, and decisions and outcomes were assessed. RESULTS Study outcomes were available for 64 patients. The mean number of genes altered per tumor was 4.97 (median=4; range, 1-26), and the average turnaround time from testing laboratory report to generation of formal recommendations was approximately three weeks. Evaluation of genomic and clinical data by the MTB led to generation of targeted treatment options in all 64 patients, and the percentage of patients for whom one or more of these recommendations were implemented by the treating physician was 39%. Sixty-four percent of the patients receiving targeted therapy based on a CGP result experienced radiologic response or showed evidence of clinical benefit or stable disease. CONCLUSION These data suggest that an institutional MTB is a feasible venue for reviewing tumor genomic profiling results and generating clinical recommendations. These data also support the need for further studies and guidelines on clinical decision making with greater availability of broad genomically based diagnostics.
Collapse
Affiliation(s)
| | - Kim M Hirshfield
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Veronica Rojas
- Rutgers Robert Wood Johnson Medical School, Rutgers University, 671 Hoes Lane, Piscataway, NJ 08854, USA
| | - Robert S DiPaola
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Darlene Gibbon
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Mira Hellmann
- Hackensack University Medical Center, John Theurer Cancer Center, 92 2nd Street, Hackensack, NJ, 07601, USA
| | - Sara Isani
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Aliza Leiser
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Gregory M Riedlinger
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Allison Wagreich
- Morristown Medical Center, Atlantic Health System, 100 Madison Avenue, Morristown, NJ 07960, USA
| | - Siraj M Ali
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA 02141, USA
| | - Julia A Elvin
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA 02141, USA
| | - Vincent A Miller
- Foundation Medicine, Inc., 150 Second Street, Cambridge, MA 02141, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| |
Collapse
|
48
|
Xu J, Pham CG, Albanese SK, Dong Y, Oyama T, Lee CH, Rodrik-Outmezguine V, Yao Z, Han S, Chen D, Parton DL, Chodera JD, Rosen N, Cheng EH, Hsieh JJ. Mechanistically distinct cancer-associated mTOR activation clusters predict sensitivity to rapamycin. J Clin Invest 2016; 126:3526-40. [PMID: 27482884 DOI: 10.1172/jci86120] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/02/2016] [Indexed: 12/21/2022] Open
Abstract
Genomic studies have linked mTORC1 pathway-activating mutations with exceptional response to treatment with allosteric inhibitors of mTORC1 called rapalogs. Rapalogs are approved for selected cancer types, including kidney and breast cancers. Here, we used sequencing data from 22 human kidney cancer cases to identify the activating mechanisms conferred by mTOR mutations observed in human cancers and advance precision therapeutics. mTOR mutations that clustered in focal adhesion kinase targeting domain (FAT) and kinase domains enhanced mTORC1 kinase activity, decreased nutrient reliance, and increased cell size. We identified 3 distinct mechanisms of hyperactivation, including reduced binding to DEP domain-containing MTOR-interacting protein (DEPTOR), resistance to regulatory associated protein of mTOR-mediated (RAPTOR-mediated) suppression, and altered kinase kinetics. Of the 28 mTOR double mutants, activating mutations could be divided into 6 complementation groups, resulting in synergistic Rag- and Ras homolog enriched in brain-independent (RHEB-independent) mTORC1 activation. mTOR mutants were resistant to DNA damage-inducible transcript 1-mediated (REDD1-mediated) inhibition, confirming that activating mutations can bypass the negative feedback pathway formed between HIF1 and mTORC1 in the absence of von Hippel-Lindau (VHL) tumor suppressor expression. Moreover, VHL-deficient cells that expressed activating mTOR mutants grew tumors that were sensitive to rapamycin treatment. These data may explain the high incidence of mTOR mutations observed in clear cell kidney cancer, where VHL loss and HIF activation is pathognomonic. Our study provides mechanistic and therapeutic insights concerning mTOR mutations in human diseases.
Collapse
|
49
|
Orienti I, Falconi M, Teti G, Currier MA, Wang J, Phelps M, Cripe TP. Preparation and Evaluation of a Novel Class of Amphiphilic Amines as Antitumor Agents and Nanocarriers for Bioactive Molecules. Pharm Res 2016; 33:2722-35. [PMID: 27457066 PMCID: PMC5040747 DOI: 10.1007/s11095-016-1999-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/11/2016] [Indexed: 12/15/2022]
Abstract
Purpose We describe a novel class of antitumor amphiphilic amines (RCn) based on a tricyclic amine hydrophilic head and a hydrophobic linear alkyl tail of variable length. Methods We tested the lead compound, RC16, for cytotoxicity and mechanism of cell death in several cancer cell lines, anti tumor efficacy in mouse tumor models, and ability to encapsulate chemotherapy drugs. Results These compounds displayed strong cytotoxic activity against cell lines derived from both pediatric and adult cancers. The IC50 of the lead compound, RC16, for normal cells including human keratinocytes, human fibroblasts and human umbilical vein endothelial cells was tenfold higher than for tumor cells. RC16 exhibited significant antitumor effects in vivo using several human xenografts and a metastatic model of murine neuroblastoma by both intravenous and oral administration routes. The amphiphilic character of RC16 triggered a spontaneous molecular self-assembling in water with formation of micelles allowing complexation of Doxorubicin, Etoposide and Paclitaxel. These micelles significantly improved the in vitro antitumor activity of these drugs as the enhancement of their aqueous solubility also improved their biologic availability. Conclusions RC16 and related amphiphilic amines may be useful as a novel cancer treatment. Electronic supplementary material The online version of this article (doi:10.1007/s11095-016-1999-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Isabella Orienti
- Department of Pharmacy and Biotechnology, University of Bologna, Via S. Donato 19/2, Bologna, Italy
| | - Mirella Falconi
- Department for Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, via Irnerio 48, Bologna, Italy
| | - Gabriella Teti
- Department for Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, via Irnerio 48, Bologna, Italy
| | - Mark A Currier
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jiang Wang
- College of Pharmacy and Division of Pharmaceutics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, 43210, USA
| | - Mitch Phelps
- College of Pharmacy and Division of Pharmaceutics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, 43210, USA
| | - Timothy P Cripe
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.
- Division of Hematology/Oncology/Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Dr, Columbus, Ohio, 43205, USA.
| |
Collapse
|
50
|
Rioth MJ, Thota R, Staggs DB, Johnson DB, Warner JL. Pragmatic precision oncology: the secondary uses of clinical tumor molecular profiling. J Am Med Inform Assoc 2016; 23:773-6. [PMID: 27026612 PMCID: PMC4926741 DOI: 10.1093/jamia/ocw002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/07/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Precision oncology increasingly utilizes molecular profiling of tumors to determine treatment decisions with targeted therapeutics. The molecular profiling data is valuable in the treatment of individual patients as well as for multiple secondary uses. OBJECTIVE To automatically parse, categorize, and aggregate clinical molecular profile data generated during cancer care as well as use this data to address multiple secondary use cases. METHODS A system to parse, categorize and aggregate molecular profile data was created. A naÿve Bayesian classifier categorized results according to clinical groups. The accuracy of these systems were validated against a published expertly-curated subset of molecular profiling data. RESULTS Following one year of operation, 819 samples have been accurately parsed and categorized to generate a data repository of 10,620 genetic variants. The database has been used for operational, clinical trial, and discovery science research. CONCLUSIONS A real-time database of molecular profiling data is a pragmatic solution to several knowledge management problems in the practice and science of precision oncology.
Collapse
Affiliation(s)
- Matthew J Rioth
- Department of Medicine, Division of Hematology & Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232 Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, 37203 USA
| | - Ramya Thota
- Department of Medicine, Division of Hematology & Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - David B Staggs
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, 37203 USA
| | - Douglas B Johnson
- Department of Medicine, Division of Hematology & Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Jeremy L Warner
- Department of Medicine, Division of Hematology & Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232,Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, 37203 USA
| |
Collapse
|