1
|
Trin K, Dalleau C, Mathoulin-Pelissier S, Le Tourneau C, Dinart D, Bellera C. The Growth Modulation Index (GMI) as an Efficacy Outcome in Cancer Clinical Trials: A Scoping Review with Suggested Reporting Guidelines. Curr Oncol Rep 2025; 27:516-532. [PMID: 40156702 DOI: 10.1007/s11912-025-01667-1] [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] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE OF REVIEW The growth modulation index (GMI) is defined as the ratio between the time to progression of a new line of treatment and the previous line. This ratio can be used to determine whether the new line of treatment brings a clinical benefit. It has been proposed as an outcome in trials evaluating non-cytotoxic drugs. Its interest lies in the intra-patient comparison. The terminology employed to refer to the GMI, as well as its definitions, are highly variable in the literature. Some uses of the GMI are arbitrary and not based on any scientific rationale. Our aim is to describe how the GMI is reported in the scientific literature. RECENT FINDINGS We carried out a scoping review using PubMed, Scopus, Web of Science and BASE (Bielefeld Academic Search Engine). The algorithm was composed of the terms "growth modulation index", "time to progression ratio" and "progression-free survival ratio". Documents in English, with full-text available, published up to 2023, were included. Among 227 included documents, 166 of which discussed GMI specifically. On these 166 documents, 76 reported on observational studies, 62 on interventional studies and 17 on methodological or statistical developments pertaining to the GMI. All were about oncology. Our review highlights significant variability in the reporting and use of the GMI. To address this, we propose standardized reporting guidelines. Additionally, we emphasize the need for methodological and statistical developments to improve the use of the GMI and to develop novel GMI-based trial designs.
Collapse
Affiliation(s)
- Kilian Trin
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France.
- Medical Science Faculty, University of Bordeaux, Bordeaux, France.
| | - Cynthia Dalleau
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| | - Simone Mathoulin-Pelissier
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- INSERM U900 Research Unit, Institut Curie, Paris, France
- Paris-Saclay University, Paris, France
| | - Derek Dinart
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| | - Carine Bellera
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| |
Collapse
|
2
|
Abstract
Adenosquamous carcinoma (ASC), containing both adenocarcinoma and squamous cell carcinoma components, is rare in the digestive system. Limited data is available on ASC of the digestive system (AS-ASC), and the current evidence is available mainly in the form of case reports and case series. We performed a thorough search of the available literature and compiled a review on the epidemiology, histopathology, pathogenesis, clinical manifestations, diagnosis, treatment, and prognosis of AS-ASC. Non-specific clinical and imaging presentations and low diagnostic accuracy of biopsy lead to difficulties in preoperative diagnosis in a high proportion of patients and high malignancy. The pathogenesis remains obscure. Surgery remains the mainstay of treatment for AS-ASC. The role of chemoradiotherapy as an adjuvant treatment is still inconclusive. Key messages Metastatic linings and the lack of efficacious treatments lead to an unfavorable outcome in AS-ASC patients. Further research could help us understand the pathophysiology of AS-ASCand the unique needs of AS-ASC patients.
Collapse
Affiliation(s)
- Hong-Shuai Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Tao He
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Li-Li Yang
- Department of Medical Oncology, Chengdu Shangjinnanfu Hospital, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Gately S. Human Microbiota and Personalized Cancer Treatments: Role of Commensal Microbes in Treatment Outcomes for Cancer Patients. Cancer Treat Res 2019; 178:253-264. [PMID: 31209849 DOI: 10.1007/978-3-030-16391-4_10] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The human gut microbiota consists of about 3.8 × 1013 microorganisms that play an essential role in health, metabolism, and immunomodulation. These gut microbes alter therapeutic response and toxicity to cancer therapies including cytotoxic chemotherapy, radiation therapy, kinase inhibitors, and immunotherapy agents. The gut microbiota generates short-chain fatty acids that are significant regulators of histone post-translational modifications that fundamentally regulate gene expression, linking the microbiota to cellular metabolism and transcriptional regulation. The short-chain fatty acids not only act locally but can be taken up in the blood stream to inhibit the activity of histone deacetylases, regulate gene expression in distant organs as well as the effector function of CD8+ T cells. Cancer and the treatments for it negatively impact the microbiome often resulting in dysbiosis. This can diminish a patient's response to treatment as well as increase systemic toxicities from these therapies. In addition to the gut microbiota, microbes have been detected in tumors that can modulate chemotherapeutic drug response and can result in immune suppression. The gut microbiota and tumor-associated bacteria may be a significant contributor to the interindividual differences and heterogeneous responses to cancer therapies and drug tolerability and strategies that support and/or manipulate the microbiota to improve therapeutic outcome is an emerging area for personalized cancer treatment.
Collapse
Affiliation(s)
- Stephen Gately
- Translational Drug Development (TD2), Scottsdale, AZ, USA.
| |
Collapse
|
4
|
Powell SF, Dib EG, Bleeker JS, Keppen MD, Mazurczak M, Hack KM, Gitau MM, Steen PD, Terstriep SA, Reynolds J, Landsverk ML, Chan CH, Nelson ME, Thompson PA, Ellison C, Black LJ, Ford JM, Chung JH, Anhorn R, Gaba AG. Delivering Precision Oncology in a Community Cancer Program: Results From a Prospective Observational Study. JCO Precis Oncol 2018; 2:1-12. [DOI: 10.1200/po.17.00220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Introduction Precision oncology (PO) is a growing treatment approach in the era of next-generation sequencing (NGS) and matched therapies. Effective delivery of PO in the community has not been extensively studied. Our program developed a virtual molecular tumor board (MTB) strategy to help guide PO care. Materials and Methods Over 18 months, eligible adult patients with advanced, incurable solid tumor malignancies were enrolled in a molecular profiling (MP) study using the Foundation Medicine NGS panel. Results were reviewed through a weekly, videoconferenced MTB conducted across our largely rural integrated health system. Recommendations from the MTB were used to identify actionable alterations (AAs). Feasibility of PO care delivery was assessed as the primary outcome. Secondary outcomes included the frequency of AAs, genomic matched treatments, genomic matched clinical trial enrollment, and clinical outcomes. Results A total of 120 participants with a variety of advanced tumor types were enrolled. Of these, 109 (90.8%) had successful MP. Treatment on the basis of an AA was recommended by the MTB in 58% of patients (63 of 109) who had a successful MP result. For those completing MP, treatments included enrollment in a genomic matched clinical trial (n = 16; 14.6%) and genomic matched treatment with a Food and Drug Administration–approved agent (n = 23; 21.1%). Response and survival data were similar regardless of the matched treatment option chosen. Conclusion A video-conferenced MTB-facilitated NGS testing and treatment delivery system was implemented in our integrated community oncology program. Continued use of this model aims to increase understanding of the impact of PO in this setting.
Collapse
Affiliation(s)
- Steven F. Powell
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Elie G. Dib
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Jonathan S. Bleeker
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Michael D. Keppen
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Miroslaw Mazurczak
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Keely M. Hack
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Mark M. Gitau
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Preston D. Steen
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Shelby A. Terstriep
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - John Reynolds
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Megan L. Landsverk
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Chun-Hung Chan
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Morgan E. Nelson
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Paul A. Thompson
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Christie Ellison
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Lora J. Black
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - James M. Ford
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Jon H. Chung
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Rachel Anhorn
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| | - Anu G. Gaba
- Steven F. Powell, Elie G. Dib, Jonathan S. Bleeker, Michael D. Keppen, Miroslaw Mazurczak, Keely M. Hack, Megan L. Landsverk, and Chun-Hung Chan, Sanford Cancer Center; Morgan E. Nelson, Paul A. Thompson, Christie Ellison, and Lora J. Black, Sanford Research, Sioux Falls, SD; Mark M. Gitau, Preston D. Steen, Shelby A. Terstriep, and Anu G. Gaba, Roger Maris Cancer Center, Fargo; John Reynolds, Sanford Cancer Center, Bismarck, ND; James M. Ford, Stanford University School of Medicine, Stanford, CA; Jon H
| |
Collapse
|
5
|
Clinical cancer genomic profiling by three-platform sequencing of whole genome, whole exome and transcriptome. Nat Commun 2018; 9:3962. [PMID: 30262806 PMCID: PMC6160438 DOI: 10.1038/s41467-018-06485-7] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/24/2018] [Indexed: 12/17/2022] Open
Abstract
To evaluate the potential of an integrated clinical test to detect diverse classes of somatic and germline mutations relevant to pediatric oncology, we performed three-platform whole-genome (WGS), whole exome (WES) and transcriptome (RNA-Seq) sequencing of tumors and normal tissue from 78 pediatric cancer patients in a CLIA-certified, CAP-accredited laboratory. Our analysis pipeline achieves high accuracy by cross-validating variants between sequencing types, thereby removing the need for confirmatory testing, and facilitates comprehensive reporting in a clinically-relevant timeframe. Three-platform sequencing has a positive predictive value of 97–99, 99, and 91% for somatic SNVs, indels and structural variations, respectively, based on independent experimental verification of 15,225 variants. We report 240 pathogenic variants across all cases, including 84 of 86 known from previous diagnostic testing (98% sensitivity). Combined WES and RNA-Seq, the current standard for precision oncology, achieved only 78% sensitivity. These results emphasize the critical need for incorporating WGS in pediatric oncology testing. Clinical oncology is rapidly adopting next-generation sequencing technology for nucleotide variant and indel detection. Here the authors present a three-platform approach (whole-genome, whole-exome, and whole-transcriptome) in pediatric patients for the detection of diverse types of germline and somatic variants.
Collapse
|
6
|
Gong J, Pan K, Fakih M, Pal S, Salgia R. Value-based genomics. Oncotarget 2018; 9:15792-15815. [PMID: 29644010 PMCID: PMC5884665 DOI: 10.18632/oncotarget.24353] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/19/2018] [Indexed: 12/18/2022] Open
Abstract
Advancements in next-generation sequencing have greatly enhanced the development of biomarker-driven cancer therapies. The affordability and availability of next-generation sequencers have allowed for the commercialization of next-generation sequencing platforms that have found widespread use for clinical-decision making and research purposes. Despite the greater availability of tumor molecular profiling by next-generation sequencing at our doorsteps, the achievement of value-based care, or improving patient outcomes while reducing overall costs or risks, in the era of precision oncology remains a looming challenge. In this review, we highlight available data through a pre-established and conceptualized framework for evaluating value-based medicine to assess the cost (efficiency), clinical benefit (effectiveness), and toxicity (safety) of genomic profiling in cancer care. We also provide perspectives on future directions of next-generation sequencing from targeted panels to whole-exome or whole-genome sequencing and describe potential strategies needed to attain value-based genomics.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Kathy Pan
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marwan Fakih
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Sumanta Pal
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Ravi Salgia
- Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
7
|
Weiss GJ, Byron SA, Aldrich J, Sangal A, Barilla H, Kiefer JA, Carpten JD, Craig DW, Whitsett TG. A prospective pilot study of genome-wide exome and transcriptome profiling in patients with small cell lung cancer progressing after first-line therapy. PLoS One 2017; 12:e0179170. [PMID: 28586388 PMCID: PMC5460863 DOI: 10.1371/journal.pone.0179170] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/19/2017] [Indexed: 11/19/2022] Open
Abstract
Background Small cell lung cancer (SCLC) that has progressed after first-line therapy is an aggressive disease with few effective therapeutic strategies. In this prospective study, we employed next-generation sequencing (NGS) to identify therapeutically actionable alterations to guide treatment for advanced SCLC patients. Methods Twelve patients with SCLC were enrolled after failing platinum-based chemotherapy. Following informed consent, genome-wide exome and RNA-sequencing was performed in a CLIA-certified, CAP-accredited environment. Actionable targets were identified and therapeutic recommendations made from a pharmacopeia of FDA-approved drugs. Clinical response to genomically-guided treatment was evaluated by Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. Results The study completed its accrual goal of 12 evaluable patients. The minimum tumor content for successful NGS was 20%, with a median turnaround time from sample collection to genomics-based treatment recommendation of 27 days. At least two clinically actionable targets were identified in each patient, and six patients (50%) received treatment identified by NGS. Two had partial responses by RECIST 1.1 on a clinical trial involving a PD-1 inhibitor + irinotecan (indicated by MLH1 alteration). The remaining patients had clinical deterioration before NGS recommended therapy could be initiated. Conclusions Comprehensive genomic profiling using NGS identified clinically-actionable alterations in SCLC patients who progressed on initial therapy. Recommended PD-1 therapy generated partial responses in two patients. Earlier access to NGS guided therapy, along with improved understanding of those SCLC patients likely to respond to immune-based therapies, should help to extend survival in these cases with poor outcomes.
Collapse
Affiliation(s)
- Glen J. Weiss
- Western Regional Medical Center, Cancer Treatment Centers of America, Goodyear, Arizona, United States of America
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
- * E-mail:
| | - Sara A. Byron
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jessica Aldrich
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Ashish Sangal
- Western Regional Medical Center, Cancer Treatment Centers of America, Goodyear, Arizona, United States of America
| | - Heather Barilla
- Western Regional Medical Center, Cancer Treatment Centers of America, Goodyear, Arizona, United States of America
| | - Jeffrey A. Kiefer
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - John D. Carpten
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - David W. Craig
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Timothy G. Whitsett
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| |
Collapse
|
8
|
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
|
9
|
Zhang X, Han C, He J. Recent Advances in the Diagnosis and Management of Bladder Cancer. Cell Biochem Biophys 2017; 73:11-5. [PMID: 25716337 DOI: 10.1007/s12013-015-0632-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The most common malignancy of urinary tract is bladder cancer. It is one of the most widespread cancers of the world and ranks nine among frequent malignancies existing in world. The only solution to above burning problem is timely diagnosis at earlier stage, and the cancer research is being forwarded in this direction. There are various prominent gene modifications responsible for growth of bladder cancer. The present review is focused on recent advances in the field of cancer makers involving, genetic, urinary, pathological, etc., approaches to contain the deadly process of carcinogenesis. The present review provides an insight on the emerging biomarkers that could be developed to boost current bladder cancer detection strategies. This shall help timely diagnosis of this deadly disease at early stage, thereby, helping bladder cancer patients to fight against this iniquity of life.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Conghui Han
- Department of Urology Surgery, Xuzhou Central Hospital, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Jantai He
- Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
10
|
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: 2.2] [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
|
11
|
Next-Generation Sequencing in Cancer Diagnostics. J Mol Diagn 2016; 18:813-816. [PMID: 27664753 DOI: 10.1016/j.jmoldx.2016.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 08/23/2016] [Indexed: 12/14/2022] Open
Abstract
This commentary highlights the article by Misyura et al that underscores the use of next-generation sequencing platforms for detection and verification of somatic variants.
Collapse
|
12
|
Bhat A, Mokou M, Zoidakis J, Jankowski V, Vlahou A, Mischak H. BcCluster: A Bladder Cancer Database at the Molecular Level. Bladder Cancer 2016; 2:65-76. [PMID: 27376128 PMCID: PMC4927921 DOI: 10.3233/blc-150024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Bladder Cancer (BC) has two clearly distinct phenotypes. Non-muscle invasive BC has good prognosis and is treated with tumor resection and intravesical therapy whereas muscle invasive BC has poor prognosis and requires usually systemic cisplatin based chemotherapy either prior to or after radical cystectomy. Neoadjuvant chemotherapy is not often used for patients undergoing cystectomy. High-throughput analytical omics techniques are now available that allow the identification of individual molecular signatures to characterize the invasive phenotype. However, a large amount of data produced by omics experiments is not easily accessible since it is often scattered over many publications or stored in supplementary files. OBJECTIVE To develop a novel open-source database, BcCluster (http://www.bccluster.org/), dedicated to the comprehensive molecular characterization of muscle invasive bladder carcinoma. MATERIALS A database was created containing all reported molecular features significant in invasive BC. The query interface was developed in Ruby programming language (version 1.9.3) using the web-framework Rails (version 4.1.5) (http://rubyonrails.org/). RESULTS BcCluster contains the data from 112 published references, providing 1,559 statistically significant features relative to BC invasion. The database also holds 435 protein-protein interaction data and 92 molecular pathways significant in BC invasion. The database can be used to retrieve binding partners and pathways for any protein of interest. We illustrate this possibility using survivin, a known BC biomarker. CONCLUSIONS BcCluster is an online database for retrieving molecular signatures relative to BC invasion. This application offers a comprehensive view of BC invasiveness at the molecular level and allows formulation of research hypotheses relevant to this phenotype.
Collapse
Affiliation(s)
- Akshay Bhat
- Charité-Universitätsmedizin Berlin, Berlin, Germany; Mosaiques diagnostics GmbH, Hannover, Germany
| | - Marika Mokou
- Biomedical Research Foundation Academy of Athens , Biotechnology Division, Athens, Greece
| | - Jerome Zoidakis
- Biomedical Research Foundation Academy of Athens , Biotechnology Division, Athens, Greece
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR) , Aachen, Germany
| | - Antonia Vlahou
- Biomedical Research Foundation Academy of Athens , Biotechnology Division, Athens, Greece
| | - Harald Mischak
- Mosaiques diagnostics GmbH, Hannover, Germany; BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| |
Collapse
|
13
|
Block KI, Gyllenhaal C, Lowe L, Amedei A, Amin ARMR, Amin A, Aquilano K, Arbiser J, Arreola A, Arzumanyan A, Ashraf SS, Azmi AS, Benencia F, Bhakta D, Bilsland A, Bishayee A, Blain SW, Block PB, Boosani CS, Carey TE, Carnero A, Carotenuto M, Casey SC, Chakrabarti M, Chaturvedi R, Chen GZ, Chen H, Chen S, Chen YC, Choi BK, Ciriolo MR, Coley HM, Collins AR, Connell M, Crawford S, Curran CS, Dabrosin C, Damia G, Dasgupta S, DeBerardinis RJ, Decker WK, Dhawan P, Diehl AME, Dong JT, Dou QP, Drew JE, Elkord E, El-Rayes B, Feitelson MA, Felsher DW, Ferguson LR, Fimognari C, Firestone GL, Frezza C, Fujii H, Fuster MM, Generali D, Georgakilas AG, Gieseler F, Gilbertson M, Green MF, Grue B, Guha G, Halicka D, Helferich WG, Heneberg P, Hentosh P, Hirschey MD, Hofseth LJ, Holcombe RF, Honoki K, Hsu HY, Huang GS, Jensen LD, Jiang WG, Jones LW, Karpowicz PA, Keith WN, Kerkar SP, Khan GN, Khatami M, Ko YH, Kucuk O, Kulathinal RJ, Kumar NB, Kwon BS, Le A, Lea MA, Lee HY, Lichtor T, Lin LT, Locasale JW, Lokeshwar BL, Longo VD, Lyssiotis CA, MacKenzie KL, Malhotra M, Marino M, Martinez-Chantar ML, Matheu A, et alBlock KI, Gyllenhaal C, Lowe L, Amedei A, Amin ARMR, Amin A, Aquilano K, Arbiser J, Arreola A, Arzumanyan A, Ashraf SS, Azmi AS, Benencia F, Bhakta D, Bilsland A, Bishayee A, Blain SW, Block PB, Boosani CS, Carey TE, Carnero A, Carotenuto M, Casey SC, Chakrabarti M, Chaturvedi R, Chen GZ, Chen H, Chen S, Chen YC, Choi BK, Ciriolo MR, Coley HM, Collins AR, Connell M, Crawford S, Curran CS, Dabrosin C, Damia G, Dasgupta S, DeBerardinis RJ, Decker WK, Dhawan P, Diehl AME, Dong JT, Dou QP, Drew JE, Elkord E, El-Rayes B, Feitelson MA, Felsher DW, Ferguson LR, Fimognari C, Firestone GL, Frezza C, Fujii H, Fuster MM, Generali D, Georgakilas AG, Gieseler F, Gilbertson M, Green MF, Grue B, Guha G, Halicka D, Helferich WG, Heneberg P, Hentosh P, Hirschey MD, Hofseth LJ, Holcombe RF, Honoki K, Hsu HY, Huang GS, Jensen LD, Jiang WG, Jones LW, Karpowicz PA, Keith WN, Kerkar SP, Khan GN, Khatami M, Ko YH, Kucuk O, Kulathinal RJ, Kumar NB, Kwon BS, Le A, Lea MA, Lee HY, Lichtor T, Lin LT, Locasale JW, Lokeshwar BL, Longo VD, Lyssiotis CA, MacKenzie KL, Malhotra M, Marino M, Martinez-Chantar ML, Matheu A, Maxwell C, McDonnell E, Meeker AK, Mehrmohamadi M, Mehta K, Michelotti GA, Mohammad RM, Mohammed SI, Morre DJ, Muralidhar V, Muqbil I, Murphy MP, Nagaraju GP, Nahta R, Niccolai E, Nowsheen S, Panis C, Pantano F, Parslow VR, Pawelec G, Pedersen PL, Poore B, Poudyal D, Prakash S, Prince M, Raffaghello L, Rathmell JC, Rathmell WK, Ray SK, Reichrath J, Rezazadeh S, Ribatti D, Ricciardiello L, Robey RB, Rodier F, Rupasinghe HPV, Russo GL, Ryan EP, Samadi AK, Sanchez-Garcia I, Sanders AJ, Santini D, Sarkar M, Sasada T, Saxena NK, Shackelford RE, Shantha Kumara HMC, Sharma D, Shin DM, Sidransky D, Siegelin MD, Signori E, Singh N, Sivanand S, Sliva D, Smythe C, Spagnuolo C, Stafforini DM, Stagg J, Subbarayan PR, Sundin T, Talib WH, Thompson SK, Tran PT, Ungefroren H, Vander Heiden MG, Venkateswaran V, Vinay DS, Vlachostergios PJ, Wang Z, Wellen KE, Whelan RL, Yang ES, Yang H, Yang X, Yaswen P, Yedjou C, Yin X, Zhu J, Zollo M. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin Cancer Biol 2015; 35 Suppl:S276-S304. [PMID: 26590477 PMCID: PMC4819002 DOI: 10.1016/j.semcancer.2015.09.007] [Show More Authors] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 08/12/2015] [Accepted: 09/14/2015] [Indexed: 12/14/2022]
Abstract
Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.
Collapse
Affiliation(s)
- Keith I Block
- Block Center for Integrative Cancer Treatment, Skokie, IL, United States.
| | | | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada; Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster, United Kingdom.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - A R M Ruhul Amin
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Jack Arbiser
- Winship Cancer Institute of Emory University, Atlanta, GA, United States; Atlanta Veterans Administration Medical Center, Atlanta, GA, United States; Department of Dermatology, Emory University School of Medicine, Emory University, Atlanta, GA, United States
| | - Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Penny B Block
- Block Center for Integrative Cancer Treatment, Skokie, IL, United States
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Thomas E Carey
- Head and Neck Cancer Biology Laboratory, University of Michigan, Ann Arbor, MI, United States
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Marianeve Carotenuto
- Centro di Ingegneria Genetica e Biotecnologia Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Stephanie C Casey
- Stanford University, Division of Oncology, Department of Medicine and Pathology, Stanford, CA, United States
| | - Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, School of Medicine, Columbia, SC, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Georgia Zhuo Chen
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Helen Chen
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | - Yi Charlie Chen
- Department of Biology, Alderson Broaddus University, Philippi, WV, United States
| | - Beom K Choi
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | | | - Helen M Coley
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Andrew R Collins
- Department of Nutrition, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Sarah Crawford
- Cancer Biology Research Laboratory, Southern Connecticut State University, New Haven, CT, United States
| | - Colleen S Curran
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Charlotta Dabrosin
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Giovanna Damia
- Department of Oncology, Istituto Di Ricovero e Cura a Carattere Scientifico - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Santanu Dasgupta
- Department of Cellular and Molecular Biology, the University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas - Southwestern Medical Center, Dallas, TX, United States
| | - William K Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Punita Dhawan
- Department of Surgery and Cancer Biology, Division of Surgical Oncology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Anna Mae E Diehl
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Jin-Tang Dong
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Q Ping Dou
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Janice E Drew
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Eyad Elkord
- College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Dean W Felsher
- Stanford University, Division of Oncology, Department of Medicine and Pathology, Stanford, CA, United States
| | - Lynnette R Ferguson
- Discipline of Nutrition and Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Gary L Firestone
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Mark M Fuster
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, CA, United States
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy; Molecular Therapy and Pharmacogenomics Unit, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Frank Gieseler
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | - Michelle F Green
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Brendan Grue
- Departments of Environmental Science, Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, United States
| | | | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Patricia Hentosh
- School of Medical Laboratory and Radiation Sciences, Old Dominion University, Norfolk, VA, United States
| | - Matthew D Hirschey
- Department of Medicine, Duke University Medical Center, Durham, NC, United States; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Lorne J Hofseth
- College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Gloria S Huang
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
| | - Lasse D Jensen
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wen G Jiang
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Lee W Jones
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States
| | | | | | - Sid P Kerkar
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | | | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (Retired), National Institutes of Health, Bethesda, MD, United States
| | - Young H Ko
- University of Maryland BioPark, Innovation Center, KoDiscovery, Baltimore, MD, United States
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Nagi B Kumar
- Moffitt Cancer Center, University of South Florida College of Medicine, Tampa, FL, United States
| | - Byoung S Kwon
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi, Republic of Korea; Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Anne Le
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael A Lea
- New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Ho-Young Lee
- College of Pharmacy, Seoul National University, South Korea
| | - Terry Lichtor
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jason W Locasale
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Bal L Lokeshwar
- Department of Medicine, Georgia Regents University Cancer Center, Augusta, GA, United States
| | - Valter D Longo
- Andrus Gerontology Center, Division of Biogerontology, University of Southern California, Los Angeles, CA, United States
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology and Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, United States
| | - Karen L MacKenzie
- Children's Cancer Institute Australia, Kensington, New South Wales, Australia
| | - Meenakshi Malhotra
- Department of Biomedical Engineering, McGill University, Montréal, Canada
| | - Maria Marino
- Department of Science, University Roma Tre, Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | | | - Christopher Maxwell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Eoin McDonnell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mahya Mehrmohamadi
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gregory A Michelotti
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - D James Morre
- Mor-NuCo, Inc, Purdue Research Park, West Lafayette, IN, United States
| | - Vinayak Muralidhar
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Wellcome Trust-MRC Building, Hills Road, Cambridge, United Kingdom
| | | | - Rita Nahta
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Francesco Pantano
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Virginia R Parslow
- Discipline of Nutrition and Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Graham Pawelec
- Center for Medical Research, University of Tübingen, Tübingen, Germany
| | - Peter L Pedersen
- Departments of Biological Chemistry and Oncology, Member at Large, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Brad Poore
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Deepak Poudyal
- College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Satya Prakash
- Department of Biomedical Engineering, McGill University, Montréal, Canada
| | - Mark Prince
- Department of Otolaryngology-Head and Neck, Medical School, University of Michigan, Ann Arbor, MI, United States
| | | | - Jeffrey C Rathmell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, School of Medicine, Columbia, SC, United States
| | - Jörg Reichrath
- Center for Clinical and Experimental Photodermatology, Clinic for Dermatology, Venerology and Allergology, The Saarland University Hospital, Homburg, Germany
| | - Sarallah Rezazadeh
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy & National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - R Brooks Robey
- White River Junction Veterans Affairs Medical Center, White River Junction, VT, United States; Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Francis Rodier
- Centre de Rechercher du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, Montréal, Quebec, Canada; Université de Montréal, Département de Radiologie, Radio-Oncologie et Médicine Nucléaire, Montréal, Quebec, Canada
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gian Luigi Russo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | | | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Andrew J Sanders
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Daniele Santini
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Malancha Sarkar
- Department of Biology, University of Miami, Miami, FL, United States
| | - Tetsuro Sasada
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Neeraj K Saxena
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University, Health Shreveport, Shreveport, LA, United States
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Dong M Shin
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Markus David Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States
| | - Emanuela Signori
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Sharanya Sivanand
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Sliva
- DSTest Laboratories, Purdue Research Park, Indianapolis, IN, United States
| | - Carl Smythe
- Department of Biomedical Science, Sheffield Cancer Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Carmela Spagnuolo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Pharmacie et Institut du Cancer de Montréal, Montréal, Quebec, Canada
| | - Pochi R Subbarayan
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Tabetha Sundin
- Department of Molecular Diagnostics, Sentara Healthcare, Norfolk, VA, United States
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | - Sarah K Thompson
- Department of Surgery, Royal Adelaide Hospital, Adelaide, Australia
| | - Phuoc T Tran
- Departments of Radiation Oncology & Molecular Radiation Sciences, Oncology and Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vasundara Venkateswaran
- Department of Surgery, University of Toronto, Division of Urology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Dass S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Panagiotis J Vlachostergios
- Department of Internal Medicine, New York University Lutheran Medical Center, Brooklyn, New York, NY, United States
| | - Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Huanjie Yang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| | - Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA, United States
| | - Clement Yedjou
- Department of Biology, Jackson State University, Jackson, MS, United States
| | - Xin Yin
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, CA, United States
| | - Jiyue Zhu
- Washington State University College of Pharmacy, Spokane, WA, United States
| | - Massimo Zollo
- Centro di Ingegneria Genetica e Biotecnologia Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, Federico II, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
14
|
|
15
|
Borazanci E, Millis SZ, Korn R, Han H, Whatcott CJ, Gatalica Z, Barrett MT, Cridebring D, Hoff DDV. Adenosquamous carcinoma of the pancreas: Molecular characterization of 23 patients along with a literature review. World J Gastrointest Oncol 2015; 7:132-140. [PMID: 26380056 PMCID: PMC4569590 DOI: 10.4251/wjgo.v7.i9.132] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/22/2015] [Accepted: 07/14/2015] [Indexed: 02/05/2023] Open
Abstract
Adenosquamous carcinoma of the pancreas (ASCP) is a rare entity. Like adenocarcinoma of the pancreas, overall survival is poor. Characteristics of ASCP include central tumor necrosis, along with osteoclasts and hypercalcemia. Various theories exist as to why this histological subtype exists, as normal pancreas tissue has no benign squamous epithelium. Due to the rarity of this disease, limited molecular analysis has been performed, and those reports indicate unique molecular features of ASCP. In this paper, we characterize 23 patients diagnosed with ASCP through molecular profiling using immunohistochemistry staining, fluorescent in situ hybridization, chromogenic in situ hybridization, and gene sequencing, Additionally, we provide a comprehensive literature review of what is known to date of ASCP. Molecular characterization revealed overexpression in MRP1 (80%), MGMT (79%), TOP2A (75), RRM1 (42%), TOPO1 (42%), PTEN (45%), CMET (40%), and C-KIT (10%) among others. One hundred percent of samples tested were positive for KRAS mutations. This analysis shows heretofore unsuspected leads to be considered for treatments of this rare type of exocrine pancreas cancer. Molecular profiling may be appropriate to provide maximum information regarding the patient’s tumor. Further work should be pursued to better characterize this disease.
Collapse
|
16
|
Squillace RM, Frampton GM, Stephens PJ, Ross JS, Miller VA. Comparing two assays for clinical genomic profiling: the devil is in the data. Onco Targets Ther 2015; 8:2237-42. [PMID: 26346763 PMCID: PMC4556029 DOI: 10.2147/ott.s88908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
17
|
Schwaederle M, Zhao M, Lee JJ, Eggermont AM, Schilsky RL, Mendelsohn J, Lazar V, Kurzrock R. Impact of Precision Medicine in Diverse Cancers: A Meta-Analysis of Phase II Clinical Trials. J Clin Oncol 2015; 33:3817-25. [PMID: 26304871 DOI: 10.1200/jco.2015.61.5997] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE The impact of a personalized cancer treatment strategy (ie, matching patients with drugs based on specific biomarkers) is still a matter of debate. METHODS We reviewed phase II single-agent studies (570 studies; 32,149 patients) published between January 1, 2010, and December 31, 2012 (PubMed search). Response rate (RR), progression-free survival (PFS), and overall survival (OS) were compared for arms that used a personalized strategy versus those that did not. RESULTS Multivariable analysis (both weighted multiple linear regression and random effects meta-regression) demonstrated that the personalized approach, compared with a nonpersonalized approach, consistently and independently correlated with higher median RR (31% v 10.5%, respectively; P < .001) and prolonged median PFS (5.9 v 2.7 months, respectively; P < .001) and OS (13.7 v 8.9 months, respectively; P < .001). Nonpersonalized targeted arms had poorer outcomes compared with either personalized targeted therapy or cytotoxics, with median RR of 4%, 30%, and 11.9%, respectively; median PFS of 2.6, 6.9, and 3.3 months, respectively (all P < .001); and median OS of 8.7, 15.9, and 9.4 months, respectively (all P < .05). Personalized arms using a genomic biomarker had higher median RR and prolonged median PFS and OS (all P ≤ .05) compared with personalized arms using a protein biomarker. A personalized strategy was associated with a lower treatment-related death rate than a nonpersonalized strategy (median, 1.5% v 2.3%, respectively; P < .001). CONCLUSION Comprehensive analysis of phase II, single-agent arms revealed that, across malignancies, a personalized strategy was an independent predictor of better outcomes and fewer toxic deaths. In addition, nonpersonalized targeted therapies were associated with significantly poorer outcomes than cytotoxic agents, which in turn were worse than personalized targeted therapy.
Collapse
Affiliation(s)
- Maria Schwaederle
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France.
| | - Melissa Zhao
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France
| | - J Jack Lee
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France
| | - Alexander M Eggermont
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France
| | - Richard L Schilsky
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France
| | - John Mendelsohn
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France
| | - Vladimir Lazar
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France
| | - Razelle Kurzrock
- Maria Schwaederle, Melissa Zhao, and Razelle Kurzrock, Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA; J. Jack Lee and John Mendelsohn, The University of Texas MD Anderson Cancer Center, Houston, TX; Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA; Alexander M. Eggermont and Vladimir Lazar, Institut Gustave Roussy, University Paris-Sud; and Alexander M. Eggermont, Richard L. Schilsky, John Mendelsohn, Vladimir Lazar, and Razelle Kurzrock, Worldwide Innovative Network in Personalized Cancer Medicine, Villejuif, France
| |
Collapse
|
18
|
LoRusso PM, Boerner SA, Pilat MJ, Forman KM, Zuccaro CY, Kiefer JA, Liang WS, Hunsberger S, Redman BG, Markovic SN, Sekulic A, Bryce AH, Joseph RW, Cowey CL, Fecher LA, Sosman JA, Chapman PB, Schwartz GK, Craig DW, Carpten JD, Trent JM. Pilot Trial of Selecting Molecularly Guided Therapy for Patients with Non-V600 BRAF-Mutant Metastatic Melanoma: Experience of the SU2C/MRA Melanoma Dream Team. Mol Cancer Ther 2015; 14:1962-71. [PMID: 26063764 DOI: 10.1158/1535-7163.mct-15-0153] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/21/2015] [Indexed: 01/06/2023]
Abstract
Targeted therapies and immunotherapies have led to significant improvements in the treatment of advanced cancers, including metastatic melanoma. However, new strategies are desperately needed to overcome therapeutic resistance to these agents, as well as to identify effective treatment approaches for cancer patients that fall outside major targetable mutational subtypes (e.g., non-V600 BRAF melanoma). One such strategy is to extend the paradigm of individually tailored, molecularly targeted therapy into a broader spectrum of melanoma patients, particularly those bearing tumors without commonly recognized therapeutic targets, as well as having failed or were ineligible for immunotherapy. In this nontreatment pilot study, next-generation sequencing (NGS) technologies were utilized, including whole genome and whole transcriptome sequencing, to identify molecular aberrations in patients with non-V600 BRAF metastatic melanoma. This information was then rationally matched to an appropriate clinical treatment from a defined pharmacopeia. Five patients with advanced non-V600 BRAF metastatic melanoma were enrolled. We demonstrated successful performance of the following during a clinically relevant time period: patient tumor biopsy, quality DNA/RNA extraction, DNA/RNA-based sequencing for gene expression analysis, analysis utilizing a series of data integration methodologies, report generation, and tumor board review with formulated treatment plan. Streamlining measures were conducted based on the experiences of enrolling, collecting specimens, and analyzing the molecular signatures of patients. We demonstrated the feasibility of using NGS to identify molecular aberrations and generate an individualized treatment plan in this patient population. A randomized treatment study utilizing lessons learned from the conduct of this pilot study is currently underway.
Collapse
Affiliation(s)
- Patricia M LoRusso
- Yale Cancer Center, New Haven, Connecticut. Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Scott A Boerner
- Yale Cancer Center, New Haven, Connecticut. Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Mary Jo Pilat
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Karen M Forman
- Yale Cancer Center, New Haven, Connecticut. Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Clarice Y Zuccaro
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Winnie S Liang
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Sally Hunsberger
- Biometrics Research Branch, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Bruce G Redman
- University of Michigan Comprehensive Cancer Center (UMCCC), Ann Arbor, Michigan
| | | | | | | | | | - C Lance Cowey
- Charles A. Sammons Cancer Center/Baylor University Medical Center, Dallas, Texas
| | - Leslie Anne Fecher
- University of Michigan Comprehensive Cancer Center (UMCCC), Ann Arbor, Michigan
| | | | - Paul B Chapman
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | - David W Craig
- Translational Genomics Research Institute, Phoenix, Arizona
| | - John D Carpten
- Translational Genomics Research Institute, Phoenix, Arizona
| | | |
Collapse
|
19
|
Weiss GJ, Hoff BR, Whitehead RP, Sangal A, Gingrich SA, Penny RJ, Mallery DW, Morris SM, Thompson EJ, Loesch DM, Khemka V. Evaluation and comparison of two commercially available targeted next-generation sequencing platforms to assist oncology decision making. Onco Targets Ther 2015; 8:959-67. [PMID: 25960669 PMCID: PMC4423502 DOI: 10.2147/ott.s81995] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background It is widely acknowledged that there is value in examining cancers for genomic aberrations via next-generation sequencing (NGS). How commercially available NGS platforms compare with each other, and the clinical utility of the reported actionable results, are not well known. During the course of the current study, the Foundation One (F1) test generated data on a combination of somatic mutations, insertion and deletion polymorphisms, chromosomal abnormalities, and deoxyribonucleic acid (DNA) copy number changes at ~250× coverage, while the Paradigm Cancer Diagnostic (PCDx) test generated the same type of data at >5,000× coverage, plus provided messenger RNA (mRNA) expression levels. We sought to compare and evaluate paired formalin-fixed paraffin-embedded tumor tissue using these two platforms. Methods Samples from patients with advanced solid tumors were submitted to both the F1 and PCDx vendors for NGS analysis. Turnaround time (TAT) was calculated. Biomarkers were considered clinically actionable if they had a published association with treatment response in humans and were assigned to the following categories: commercially available drug (CA), clinical trial drug (CT), or neither option (hereafter referred to as “None”). Results The demographics of the 21 unique patient tumor samples included ten men and eleven women, with a median age of 56 years. Due to insufficient archival tissue from the same collection period, in one case, we used samples from different collections. PCDx reported first results faster than F1 in 20 cases. When received at both vendors on the same day, PCDx reported first results for 14 of 15 cases, with a median TAT of 9 days earlier than F1 (P<0.0001). Categorization of CA compared to CT and none significantly favored PCDx (P=0.012). Conclusion In the current analysis, commercially available NGS platforms provided clinically relevant actionable targets (CA or CT) in 47%–67% of diverse cancer types. In the samples analyzed, PCDx significantly outperformed F1 in TAT, and had statistically significant higher clinically relevant actionable targets categorized as CA.
Collapse
Affiliation(s)
- Glen J Weiss
- Cancer Treatment Centers of America, Western Regional Medical Center, Goodyear, AZ, USA
| | - Brandi R Hoff
- Cancer Treatment Centers of America, Western Regional Medical Center, Goodyear, AZ, USA
| | - Robert P Whitehead
- Cancer Treatment Centers of America, Western Regional Medical Center, Goodyear, AZ, USA
| | - Ashish Sangal
- Cancer Treatment Centers of America, Western Regional Medical Center, Goodyear, AZ, USA
| | - Susan A Gingrich
- Cancer Treatment Centers of America, Western Regional Medical Center, Goodyear, AZ, USA
| | | | | | | | | | | | - Vivek Khemka
- Cancer Treatment Centers of America, Western Regional Medical Center, Goodyear, AZ, USA
| |
Collapse
|
20
|
Royer-Bertrand B, Rivolta C. Whole genome sequencing as a means to assess pathogenic mutations in medical genetics and cancer. Cell Mol Life Sci 2015; 72:1463-71. [PMID: 25548800 PMCID: PMC11113357 DOI: 10.1007/s00018-014-1807-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 12/17/2022]
Abstract
The past decade has seen the emergence of next-generation sequencing (NGS) technologies, which have revolutionized the field of human molecular genetics. With NGS, significant portions of the human genome can now be assessed by direct sequence analysis, highlighting normal and pathological variants of our DNA. Recent advances have also allowed the sequencing of complete genomes, by a method referred to as whole genome sequencing (WGS). In this work, we review the use of WGS in medical genetics, with specific emphasis on the benefits and the disadvantages of this technique for detecting genomic alterations leading to Mendelian human diseases and to cancer.
Collapse
Affiliation(s)
- Beryl Royer-Bertrand
- Department of Medical Genetics, University of Lausanne, Rue Du Bugnon 27, 1005 Lausanne, Switzerland
| | - Carlo Rivolta
- Department of Medical Genetics, University of Lausanne, Rue Du Bugnon 27, 1005 Lausanne, Switzerland
| |
Collapse
|
21
|
Dearing KR, Weiss GJ. Translating next-generation sequencing from clinical trials to clinical practice for the treatment of advanced cancers. Per Med 2015; 12:155-162. [PMID: 29754537 DOI: 10.2217/pme.14.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Next-generation sequencing (NGS) is being applied in oncology care to identify specific molecular aberrations of patient's tumors. The use of NGS now allows for sequencing entire human genomes within a reasonable cost and practical time frames for treatment decision making. Further delineation of epigenetics, transcriptomics, metagenomics and NGS at the level of circulating tumor DNA reveal ever increasing complexity to understand these interactions and the roles they play in cancer. With the improvement in understanding the study of proteomics, it has become clear that NGS has room for innovation to someday include sequencing of proteins. Early embarkation of NGS incorporated into clinical trials has begun. Here, we review the feasibility and practicality of translating NGS from clinical trials to clinical practice.
Collapse
Affiliation(s)
- Kristen R Dearing
- Cancer Treatment Centers of America, 14200 Celebrate Life Way, Goodyear, AZ 85338, USA
| | - Glen J Weiss
- Cancer Treatment Centers of America, 14200 Celebrate Life Way, Goodyear, AZ 85338, USA.,CRAB-Clinical Trials Consortium, 1730 Minor Ave., Seattle, WA 98101, USA
| |
Collapse
|
22
|
Bhat A, Heinzel A, Mayer B, Perco P, Mühlberger I, Husi H, Merseburger AS, Zoidakis J, Vlahou A, Schanstra JP, Mischak H, Jankowski V. Protein interactome of muscle invasive bladder cancer. PLoS One 2015; 10:e0116404. [PMID: 25569276 PMCID: PMC4287622 DOI: 10.1371/journal.pone.0116404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 12/09/2014] [Indexed: 12/31/2022] Open
Abstract
Muscle invasive bladder carcinoma is a complex, multifactorial disease caused by disruptions and alterations of several molecular pathways that result in heterogeneous phenotypes and variable disease outcome. Combining this disparate knowledge may offer insights for deciphering relevant molecular processes regarding targeted therapeutic approaches guided by molecular signatures allowing improved phenotype profiling. The aim of the study is to characterize muscle invasive bladder carcinoma on a molecular level by incorporating scientific literature screening and signatures from omics profiling. Public domain omics signatures together with molecular features associated with muscle invasive bladder cancer were derived from literature mining to provide 286 unique protein-coding genes. These were integrated in a protein-interaction network to obtain a molecular functional map of the phenotype. This feature map educated on three novel disease-associated pathways with plausible involvement in bladder cancer, namely Regulation of actin cytoskeleton, Neurotrophin signalling pathway and Endocytosis. Systematic integration approaches allow to study the molecular context of individual features reported as associated with a clinical phenotype and could potentially help to improve the molecular mechanistic description of the disorder.
Collapse
Affiliation(s)
- Akshay Bhat
- Charité-Universitätsmedizin Berlin, Med. Klinik IV, Berlin, Germany
- Mosaiques diagnostics GmbH, Hannover, Germany
| | | | - Bernd Mayer
- emergentec biodevelopment GmbH, Vienna, Austria
| | - Paul Perco
- emergentec biodevelopment GmbH, Vienna, Austria
| | | | - Holger Husi
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Axel S. Merseburger
- Department of Urology and Urological Oncology, Hannover Medical School, Hannover, Germany
| | - Jerome Zoidakis
- Biomedical Research Foundation Academy of Athens, Biotechnology Division, Athens, Greece
| | - Antonia Vlahou
- Biomedical Research Foundation Academy of Athens, Biotechnology Division, Athens, Greece
| | - Joost P. Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Diseases, Toulouse, France
- Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Harald Mischak
- Mosaiques diagnostics GmbH, Hannover, Germany
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), Aachen, Germany
- * E-mail:
| |
Collapse
|
23
|
Hagemann IS, Devarakonda S, Lockwood CM, Spencer DH, Guebert K, Bredemeyer AJ, Al-Kateb H, Nguyen TT, Duncavage EJ, Cottrell CE, Kulkarni S, Nagarajan R, Seibert K, Baggstrom M, Waqar SN, Pfeifer JD, Morgensztern D, Govindan R. Clinical next-generation sequencing in patients with non-small cell lung cancer. Cancer 2014; 121:631-9. [DOI: 10.1002/cncr.29089] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/22/2014] [Accepted: 08/27/2014] [Indexed: 01/21/2023]
Affiliation(s)
- Ian S. Hagemann
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Siddhartha Devarakonda
- Section of Medical Oncology, Division of Hematology and Oncology, Department of Medicine; Washington University; St. Louis Missouri
| | - Christina M. Lockwood
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - David H. Spencer
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Kalin Guebert
- Section of Medical Oncology, Division of Hematology and Oncology, Department of Medicine; Washington University; St. Louis Missouri
| | - Andrew J. Bredemeyer
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Hussam Al-Kateb
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - TuDung T. Nguyen
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Eric J. Duncavage
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Catherine E. Cottrell
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Shashikant Kulkarni
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Rakesh Nagarajan
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Karen Seibert
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Maria Baggstrom
- Section of Medical Oncology, Division of Hematology and Oncology, Department of Medicine; Washington University; St. Louis Missouri
| | - Saiama N. Waqar
- Section of Medical Oncology, Division of Hematology and Oncology, Department of Medicine; Washington University; St. Louis Missouri
| | - John D. Pfeifer
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology; Washington University; St. Louis Missouri
| | - Daniel Morgensztern
- Section of Medical Oncology, Division of Hematology and Oncology, Department of Medicine; Washington University; St. Louis Missouri
| | - Ramaswamy Govindan
- Section of Medical Oncology, Division of Hematology and Oncology, Department of Medicine; Washington University; St. Louis Missouri
| |
Collapse
|
24
|
Picard C, Fischer A. Contribution of high-throughput DNA sequencing to the study of primary immunodeficiencies. Eur J Immunol 2014; 44:2854-61. [PMID: 25154746 DOI: 10.1002/eji.201444669] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 07/24/2014] [Accepted: 08/18/2014] [Indexed: 01/20/2023]
Abstract
Primary immunodeficiencies (PIDs) are inborn errors of the immune system. PIDs have been characterized immunologically for the last 60 years and genetically, principally by Sanger DNA sequencing, over the last 30 years. The advent of next-generation sequencing (NGS) in 2011, with the development of whole-exome sequencing in particular, has facilitated the identification of previously unknown genetic lesions. NGS is rapidly generating a stream of candidate variants for an increasing number of genetically undefined PIDs. The use of NGS technology is ushering in a new era, by facilitating the discovery and characterization of new PIDs in patients with infections and other phenotypes, thereby helping to improve diagnostic accuracy. This review provides a historical overview of the identification of PIDs before NGS, and the advances and limitations of the use of NGS for the diagnosis and characterization of PIDs.
Collapse
Affiliation(s)
- Capucine Picard
- Study Center for Primary Immunodeficiencies, Necker-Enfant Malades Hospital, Assistance Publique, Hôpitaux de Paris (AP-HP), Paris, France; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris, France; Imagine Institute, Sorbonne Paris Cité, Paris Descartes University, Paris, France
| | | |
Collapse
|
25
|
Ong M, Carreira S, Goodall J, Mateo J, Figueiredo I, Rodrigues DN, Perkins G, Seed G, Yap TA, Attard G, de Bono JS. Validation and utilisation of high-coverage next-generation sequencing to deliver the pharmacological audit trail. Br J Cancer 2014; 111:828-36. [PMID: 24983367 PMCID: PMC4150267 DOI: 10.1038/bjc.2014.350] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 05/14/2014] [Accepted: 06/02/2014] [Indexed: 12/22/2022] Open
Abstract
Background: Predictive biomarker development is a key challenge for novel cancer therapeutics. We explored the feasibility of next-generation sequencing (NGS) to validate exploratory genomic biomarkers that impact phase I trial selection. Methods: We prospectively enrolled 158 patients with advanced solid tumours referred for phase I clinical trials at the Royal Marsden Hospital (October 2012 to March 2013). After fresh and/or archived tumour tissue were obtained, 93 patients remained candidates for phase I trials. Results from tumour sequencing on the Illumina MiSeq were cross-validated in 27 out of 93 patients on the Ion Torrent Personal Genome Machine (IT-PGM) blinded to results. MiSeq validation with Sequenom MassARRAY OncoCarta 1.0 (Sequenom Inc., San Diego, CA, USA) was performed in a separate cohort. Results: We found 97% concordance of mutation calls by MiSeq and IT-PGM at a variant allele frequency ⩾13% and ⩾500 × depth coverage, and 91% concordance between MiSeq and Sequenom. Common ‘actionable' mutations involved deoxyribonucleic acid (DNA) repair (51%), RAS-RAF-MEK (35%), Wnt (26%), and PI3K-AKT-mTOR (24%) signalling. Out of 53, 29 (55%) patients participating in phase I trials were recommended based on identified actionable mutations. Conclusions: Targeted high-coverage NGS panels are a highly feasible single-centre technology well-suited to cross-platform validation, enrichment of trials with molecularly defined populations and hypothesis testing early in drug development.
Collapse
Affiliation(s)
- M Ong
- 1] Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK [2] Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - S Carreira
- 1] Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK [2] Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - J Goodall
- 1] Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK [2] Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - J Mateo
- 1] Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK [2] Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - I Figueiredo
- Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - D N Rodrigues
- Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - G Perkins
- Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - G Seed
- Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - T A Yap
- 1] Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK [2] Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - G Attard
- 1] Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK [2] Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - J S de Bono
- 1] Cancer Biomarkers Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK [2] Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| |
Collapse
|
26
|
Shanmugam V, Ramanathan RK, Lavender NA, Sinari S, Chadha M, Liang WS, Kurdoglu A, Izatt T, Christoforides A, Benson H, Phillips L, Baker A, Murray C, Hostetter G, Von Hoff DD, Craig DW, Carpten JD. Whole genome sequencing reveals potential targets for therapy in patients with refractory KRAS mutated metastatic colorectal cancer. BMC Med Genomics 2014; 7:36. [PMID: 24943349 PMCID: PMC4074842 DOI: 10.1186/1755-8794-7-36] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 05/29/2014] [Indexed: 02/08/2023] Open
Abstract
Background The outcome of patients with metastatic colorectal carcinoma (mCRC) following first line therapy is poor, with median survival of less than one year. The purpose of this study was to identify candidate therapeutically targetable somatic events in mCRC patient samples by whole genome sequencing (WGS), so as to obtain targeted treatment strategies for individual patients. Methods Four patients were recruited, all of whom had received > 2 prior therapy regimens. Percutaneous needle biopsies of metastases were performed with whole blood collection for the extraction of constitutional DNA. One tumor was not included in this study as the quality of tumor tissue was not sufficient for further analysis. WGS was performed using Illumina paired end chemistry on HiSeq2000 sequencing systems, which yielded coverage of greater than 30X for all samples. NGS data were processed and analyzed to detect somatic genomic alterations including point mutations, indels, copy number alterations, translocations and rearrangements. Results All 3 tumor samples had KRAS mutations, while 2 tumors contained mutations in the APC gene and the PIK3CA gene. Although we did not identify a TCF7L2-VTI1A translocation, we did detect a TCF7L2 mutation in one tumor. Among the other interesting mutated genes was INPPL1, an important gene involved in PI3 kinase signaling. Functional studies demonstrated that inhibition of INPPL1 reduced growth of CRC cells, suggesting that INPPL1 may promote growth in CRC. Conclusions Our study further supports potential molecularly defined therapeutic contexts that might provide insights into treatment strategies for refractory mCRC. New insights into the role of INPPL1 in colon tumor cell growth have also been identified. Continued development of appropriate targeted agents towards specific events may be warranted to help improve outcomes in CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - John D Carpten
- Translational Genomics Research Institute (TGen), 445 N Fifth Street, Phoenix, AZ 85004, USA.
| |
Collapse
|
27
|
Kahn SM. Next-generation sequencing for cancer drug development: the present and visions for the future. Per Med 2014; 11:139-142. [PMID: 29751374 DOI: 10.2217/pme.13.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
NGS for Cancer Drug Development 24-26 September 2013, Boston, MA, USA Many of us who, prior to the -omics revolution, staunchly pursued the molecular basis of disease causation recognized the fundamental importance that biomarkers held for the eventuality of personalized medicine, more accurate diagnostics and targeted drug discovery. With slab sequencing apparatuses now mere memories relegated to the dusty reaches of old laboratory cabinets, we are fervently unraveling the complexities of many diseases through newer, more powerful innovative technologies. Quicker than ever before, we are achieving a deeper understanding of causative genetic lesions, perturbed pathways and the functions of altered networks. This has opened the door to a golden age of data generation that shows promise as a vital key to the successful treatment of human diseases. Hanson Wade recently organized a series of three related symposia that highlighted advances in industry and academic research in the areas of drug development, companion diagnostics and disease profiling: 'NGS for Cancer Drug Development' conference in Boston on 24-26 September 2013, the 'NGS Data Analysis' conference in San Francisco on 15-17 October 2013 and the 'World CDx' conference in Boston on 12-15 November 2013. These meetings provided forums for leaders to present advancements in personalized medicine, strategies for drug development, and innovations in diagnostics and other important areas. Their highly interactive nature also provided opportunities for industry and academic attendees to identify common hurdles that stand in the way of progress, (e.g., biomarker validation, analytical software limitations, data sharing, sample handling, regulatory and reimbursement restrictions), and to propose strategies for independent and community approaches toward overcoming such hurdles. In the interest of space, this review will be limited to the first in this series, the NGS for Cancer Drug Development conference, and its six major areas of focus.
Collapse
Affiliation(s)
- Scott M Kahn
- Dept. of Urology, Columbia University, NY, USA.,Biomarkers Council, International Cancer Advocacy Network, Phoenix, AZ, USA.,Oncostem Biotherapeutics, LLC, NY, USA
| |
Collapse
|