1
|
Westphalen CB, Martins-Branco D, Beal JR, Cardone C, Coleman N, Schram AM, Halabi S, Michiels S, Yap C, André F, Bibeau F, Curigliano G, Garralda E, Kummar S, Kurzrock R, Limaye S, Loges S, Marabelle A, Marchió C, Mateo J, Rodon J, Spanic T, Pentheroudakis G, Subbiah V. The ESMO Tumour-Agnostic Classifier and Screener (ETAC-S): a tool for assessing tumour-agnostic potential of molecularly guided therapies and for steering drug development. Ann Oncol 2024; 35:936-953. [PMID: 39187421 DOI: 10.1016/j.annonc.2024.07.730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/19/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Advances in precision oncology led to approval of tumour-agnostic molecularly guided treatment options (MGTOs). The minimum requirements for claiming tumour-agnostic potential remain elusive. METHODS The European Society for Medical Oncology (ESMO) Precision Medicine Working Group (PMWG) coordinated a project to optimise tumour-agnostic drug development. International experts examined and summarised the publicly available data used for regulatory assessment of the tumour-agnostic indications approved by the US Food and Drug Administration and/or the European Medicines Agency as of December 2023. Different scenarios of minimum objective response rate (ORR), number of tumour types investigated, and number of evaluable patients per tumour type were assessed for developing a screening tool for tumour-agnostic potential. This tool was tested using the tumour-agnostic indications approved during the first half of 2024. A taxonomy for MGTOs and a framework for tumour-agnostic drug development were conceptualised. RESULTS Each tumour-agnostic indication had data establishing objective response in at least one out of five patients (ORR ≥ 20%) in two-thirds (≥4) of the investigated tumour types, with at least five evaluable patients in each tumour type. These minimum requirements were met by tested indications and may serve as a screening tool for tumour-agnostic potential, requiring further validation. We propose a conceptual taxonomy classifying MGTOs based on the therapeutic effect obtained by targeting a driver molecular aberration across tumours and its modulation by tumour-specific biology: tumour-agnostic, tumour-modulated, or tumour-restricted. The presence of biology-informed mechanistic rationale, early regulatory advice, and adequate trial design demonstrating signs of biology-driven tumour-agnostic activity, followed by confirmatory evidence, should be the principles for tumour-agnostic drug development. CONCLUSION The ESMO Tumour-Agnostic Classifier (ETAC) focuses on the interplay of targeted driver molecular aberration and tumour-specific biology modulating the therapeutic effect of MGTOs. We propose minimum requirements to screen for tumour-agnostic potential (ETAC-S) as part of tumour-agnostic drug development. Definition of ETAC cut-offs is warranted.
Collapse
Affiliation(s)
- C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich; German Cancer Consortium (DKTK), partner site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - D Martins-Branco
- Scientific and Medical Division, European Society for Medical Oncology (ESMO), Lugano, Switzerland
| | - J R Beal
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - C Cardone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Naples, Italy
| | - N Coleman
- School of Medicine, Trinity College Dublin, Dublin; Medical Oncology Department, St. James's Hospital, Dublin; Trinity St. James's Cancer Institute, Dublin, Ireland
| | - A M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City; Weill Cornell Medical College, New York City
| | - S Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham; Duke Cancer Institute, Duke University, Durham, USA
| | - S Michiels
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif, France
| | - C Yap
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - F André
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre
| | - F Bibeau
- Service d'Anatomie Pathologique, CHU Besançon, Université de Bourgogne Franche-Comté, Besançon, France
| | - G Curigliano
- Istituto Europeo di Oncologia, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - E Garralda
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - S Kummar
- Division of Hematology and Medical Oncology, Department of Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland
| | - R Kurzrock
- Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, USA
| | - S Limaye
- Medical & Precision Oncology, Sir H. N. Reliance Foundation Hospital & Research Centre, Mumbai, India
| | - S Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - A Marabelle
- Drug Development Department (DITEP) and Laboratory for Translational Research in Immunotherapy (LRTI), Gustave Roussy, INSERM U1015 & CIC1428, Université Paris-Saclay, Villejuif, France
| | - C Marchió
- Department of Medical Sciences, University of Turin, Turin; Division of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - J Rodon
- Department of Investigational Cancer Therapeutics, UT MD Anderson, Houston, USA
| | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - G Pentheroudakis
- Scientific and Medical Division, European Society for Medical Oncology (ESMO), Lugano, Switzerland
| | - V Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute (SCRI), Nashville, USA
| |
Collapse
|
2
|
Mahajan AT, Shivani, Datusalia AK, Coluccini C, Coghi P, Chaudhary S. Pyrazolo[1,5- a]pyrimidine as a Prominent Framework for Tropomyosin Receptor Kinase (Trk) Inhibitors-Synthetic Strategies and SAR Insights. Molecules 2024; 29:3560. [PMID: 39124968 PMCID: PMC11314189 DOI: 10.3390/molecules29153560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Tropomyosin receptor kinases (Trks) are transmembrane receptor tyrosine kinases named TrkA, TrkB, and TrkC and encoded by the NTRK1, NTRK2, and NTRK3 genes, respectively. These kinases have attracted significant attention and represent a promising therapeutic target for solid tumor treatment due to their vital role in cellular signaling pathways. First-generation TRK inhibitors, i.e., Larotrectinib sulfate and Entrectinib, received clinical approval in 2018 and 2019, respectively. However, the use of these inhibitors was significantly limited because of the development of resistance due to mutations. Fortunately, the second-generation Trk inhibitor Repotrectinib (TPX-0005) was approved by the FDA in November 2023, while Selitrectinib (Loxo-195) has provided an effective solution to this issue. Another macrocycle-based analog, along with many other TRK inhibitors, is currently in clinical trials. Two of the three marketed drugs for NTRK fusion cancers feature a pyrazolo[1,5-a] pyrimidine nucleus, prompting medicinal chemists to develop numerous novel pyrazolopyrimidine-based molecules to enhance clinical applications. This article focuses on a comprehensive review of chronological synthetic developments and the structure-activity relationships (SAR) of pyrazolo[1,5-a]pyrimidine derivatives as Trk inhibitors. This article will also provide comprehensive knowledge and future directions to the researchers working in the field of medicinal chemistry by facilitating the structural modification of pyrazolo [1,5-a]pyrimidine derivatives to synthesize more effective novel chemotherapeutics as TRK inhibitors.
Collapse
Affiliation(s)
- Amol T. Mahajan
- Laboratory of Bioactive Heterocycles and Catalysis (BHC Lab), Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor–Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India; (A.T.M.); (S.)
| | - Shivani
- Laboratory of Bioactive Heterocycles and Catalysis (BHC Lab), Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor–Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India; (A.T.M.); (S.)
| | - Ashok Kumar Datusalia
- Laboratory of Molecular Neurotherapeutics, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor–Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India;
| | - Carmine Coluccini
- Institute of New Drug Development, College of Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Paolo Coghi
- Laboratory for Drug Discovery from Natural Resources & Industrialization, School of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China
| | - Sandeep Chaudhary
- Laboratory of Bioactive Heterocycles and Catalysis (BHC Lab), Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor–Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India; (A.T.M.); (S.)
| |
Collapse
|
3
|
Hogervorst MA, van Hattem CC, Sonke GS, Mantel-Teeuwisse AK, Goettsch WG, Bloem LT. Healthcare decision-making for tumour-agnostic therapies in Europe: lessons learned. Drug Discov Today 2024; 29:104031. [PMID: 38796096 DOI: 10.1016/j.drudis.2024.104031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/28/2024]
Abstract
The tumour-agnostic authorisations of larotrectinib and entrectinib shifted the paradigm for indication setting. European healthcare decision-makers agreed on their therapeutic potential but diverged primarily in identified uncertainties concerning basket trial designs and endpoints, prognostic value of neurotrophic tropomyosin receptor kinase (NTRK) gene fusions, and resistance mechanisms. In addition, assessments of relevant comparators, unmet medical needs (UMNs), and implementation of NTRK-testing strategies diverged. In particular, the tumour-specific reimbursement recommendations and guidelines do not reflect tumour-agnostic thinking. These differences indicate difficulties experienced in these assessments and provide valuable lessons for future disruptive therapies. As we discuss here, early multistakeholder dialogues concerning minimum evidence requirements and involving clinicians are essential.
Collapse
Affiliation(s)
- Milou A Hogervorst
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Christine C van Hattem
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, University of Amsterdam, Amsterdam, the Netherlands
| | - Aukje K Mantel-Teeuwisse
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Wim G Goettsch
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands; National Health Care Institute (ZIN), Diemen, the Netherlands
| | - Lourens T Bloem
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
4
|
Wang X, Wei W. rBMA: A robust Bayesian Model Averaging Method for phase II basket trials based on informative mixture priors. Contemp Clin Trials 2024; 140:107505. [PMID: 38521384 DOI: 10.1016/j.cct.2024.107505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/21/2024] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Oncology drug research in the last few decades has been driven by the development of targeted agents. In the era of targeted therapies, basket trials are often used to test the antitumor activity of a novel treatment in multiple indications sharing the same genomic alteration. As patient population are further fragmented into biomarker-defined subgroups in basket trials, novel statistical methods are needed to facilitate cross-indication learning to improve the statistical power in basket trial design. Here we propose a robust Bayesian model averaging (rBMA) technique for the design and analysis of phase II basket trials. We consider the posterior distribution of each indication (basket) as the weighted average of three different models which only differ in their priors (enthusiastic, pessimistic and non-informative). The posterior weights of these models are determined based on the effect of the experimental treatment in all the indications tested. In early phase oncology trials, different binary endpoints might be chosen for different indications (objective response, disease control or PFS at landmark times), which makes it even more challenging to borrow information across indications. Compared to previous approaches, the proposed method has the flexibility to support cross-indication learning in the presence of mixed endpoints. We evaluate and compare the performance of the proposed rBMA approach to competing approaches in simulation studies. R scripts to implement the proposed method are available at https://github.com/xwang317/rBMA.
Collapse
Affiliation(s)
- Xueting Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, United States of America
| | - Wei Wei
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, United States of America.
| |
Collapse
|
5
|
Aleksakhina SN, Ivantsov AO, Imyanitov EN. Agnostic Administration of Targeted Anticancer Drugs: Looking for a Balance between Hype and Caution. Int J Mol Sci 2024; 25:4094. [PMID: 38612902 PMCID: PMC11012409 DOI: 10.3390/ijms25074094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Many tumors have well-defined vulnerabilities, thus potentially allowing highly specific and effective treatment. There is a spectrum of actionable genetic alterations which are shared across various tumor types and, therefore, can be targeted by a given drug irrespective of tumor histology. Several agnostic drug-target matches have already been approved for clinical use, e.g., immune therapy for tumors with microsatellite instability (MSI) and/or high tumor mutation burden (TMB), NTRK1-3 and RET inhibitors for cancers carrying rearrangements in these kinases, and dabrafenib plus trametinib for BRAF V600E mutated malignancies. Multiple lines of evidence suggest that this histology-independent approach is also reasonable for tumors carrying ALK and ROS1 translocations, biallelic BRCA1/2 inactivation and/or homologous recombination deficiency (HRD), strong HER2 amplification/overexpression coupled with the absence of other MAPK pathway-activating mutations, etc. On the other hand, some well-known targets are not agnostic: for example, PD-L1 expression is predictive for the efficacy of PD-L1/PD1 inhibitors only in some but not all cancer types. Unfortunately, the individual probability of finding a druggable target in a given tumor is relatively low, even with the use of comprehensive next-generation sequencing (NGS) assays. Nevertheless, the rapidly growing utilization of NGS will significantly increase the number of patients with highly unusual or exceptionally rare tumor-target combinations. Clinical trials may provide only a framework for treatment attitudes, while the decisions for individual patients usually require case-by-case consideration of the probability of deriving benefit from agnostic versus standard therapy, drug availability, associated costs, and other circumstances. The existing format of data dissemination may not be optimal for agnostic cancer medicine, as conventional scientific journals are understandably biased towards the publication of positive findings and usually discourage the submission of case reports. Despite all the limitations and concerns, histology-independent drug-target matching is certainly feasible and, therefore, will be increasingly utilized in the future.
Collapse
Affiliation(s)
- Svetlana N. Aleksakhina
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
| | - Alexander O. Ivantsov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
6
|
Hernandez S, Conde E, Molero A, Suarez-Gauthier A, Martinez R, Alonso M, Plaza C, Camacho C, Chantada D, Juaneda-Magdalena L, Garcia-Toro E, Saiz-Lopez P, Rojo F, Abad M, Boni V, Del Carmen S, Regojo RM, Sanchez-Frias ME, Teixido C, Paz-Ares L, Lopez-Rios F. Efficient Identification of Patients With NTRK Fusions Using a Supervised Tumor-Agnostic Approach. Arch Pathol Lab Med 2024; 148:318-326. [PMID: 37270803 DOI: 10.5858/arpa.2022-0443-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 06/06/2023]
Abstract
CONTEXT.— The neurotrophic tropomyosin receptor kinase (NTRK) family gene rearrangements have been recently incorporated as predictive biomarkers in a "tumor-agnostic" manner. However, the identification of these patients is extremely challenging because the overall frequency of NTRK fusions is below 1%. Academic groups and professional organizations have released recommendations on the algorithms to detect NTRK fusions. The European Society for Medical Oncology proposal encourages the use of next-generation sequencing (NGS) if available, or alternatively immunohistochemistry (IHC) could be used for screening with NGS confirmation of all positive IHC results. Other academic groups have included histologic and genomic information in the testing algorithm. OBJECTIVE.— To apply some of these triaging strategies for a more efficient identification of NTRK fusions within a single institution, so pathologists can gain practical insight on how to start looking for NTRK fusions. DESIGN.— A multiparametric strategy combining histologic (secretory carcinomas of the breast and salivary gland; papillary thyroid carcinomas; infantile fibrosarcoma) and genomic (driver-negative non-small cell lung carcinomas, microsatellite instability-high colorectal adenocarcinomas, and wild-type gastrointestinal stromal tumors) triaging was put forward. RESULTS.— Samples from 323 tumors were stained with the VENTANA pan-TRK EPR17341 Assay as a screening method. All positive IHC cases were simultaneously studied by 2 NGS tests, Oncomine Comprehensive Assay v3 and FoundationOne CDx. With this approach, the detection rate of NTRK fusions was 20 times higher (5.57%) by only screening 323 patients than the largest cohort in the literature (0.30%) comprising several hundred thousand patients. CONCLUSIONS.— Based on our findings, we propose a multiparametric strategy (ie, "supervised tumor-agnostic approach") when pathologists start searching for NTRK fusions.
Collapse
Affiliation(s)
- Susana Hernandez
- From the Department of Pathology, 12 de Octubre University Hospital, Research Institute 12 de Octubre University Hospital (i+12), Madrid, Spain (Hernandez, Alonso)
| | - Esther Conde
- the Department of Pathology, 12 de Octubre University Hospital, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain (Conde, Lopez-Rios)
| | - Aida Molero
- the Department of Pathology, Segovia General Hospital, Segovia, Spain (Molero)
| | - Ana Suarez-Gauthier
- the Department of Pathology, Jimenez Diaz Foundation University Hospital, Madrid, Spain (Suarez-Gauthier)
| | - Rebeca Martinez
- the Department of Pathology, Health Diagnostic-Grupo Quiron Salud, Madrid, Spain (Martinez)
| | - Marta Alonso
- From the Department of Pathology, 12 de Octubre University Hospital, Research Institute 12 de Octubre University Hospital (i+12), Madrid, Spain (Hernandez, Alonso)
| | - Carlos Plaza
- the Department of Pathology, Clinico San Carlos University Hospital, Madrid, Spain (Plaza)
| | - Carmen Camacho
- the Department of Pathology, Insular Materno-Infantil University Hospital, Las Palmas de Gran Canaria, Spain (Camacho)
| | - Debora Chantada
- the Department of Pathology, Alvaro Cunqueiro Hospital, Vigo, Spain (Chantada, Juaneda-Magdalena)
| | - Laura Juaneda-Magdalena
- the Department of Pathology, Alvaro Cunqueiro Hospital, Vigo, Spain (Chantada, Juaneda-Magdalena)
| | - Enrique Garcia-Toro
- the Department of Pathology, Burgos University Hospital, Burgos, Spain (Garcia-Toro, Saiz-Lopez)
| | - Patricia Saiz-Lopez
- the Department of Pathology, Burgos University Hospital, Burgos, Spain (Garcia-Toro, Saiz-Lopez)
| | - Federico Rojo
- the Institute of Health Research-Jimenez Diaz Foundation, CIBERONC, Madrid, Spain (Rojo)
| | - Mar Abad
- the Department of Pathology, Salamanca University Hospital, Salamanca, Spain (Abad)
| | - Valentina Boni
- NEXT Oncology Madrid, Quiron Salud Madrid University Hospital, Madrid, Spain (Boni)
| | - Sofia Del Carmen
- the Department of Pathology, Marques de Valdecilla University Hospital, Santander, Spain (del Carmen)
| | - Rita Maria Regojo
- the Department of Pathology, La Paz University Hospital, Madrid, Spain (Regojo)
| | | | - Cristina Teixido
- the Department of Pathology, Thoracic Oncology Unit, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Spain (Teixido)
| | - Luis Paz-Ares
- the Department of Oncology, 12 de Octubre University Hospital, Department of Medicine, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain (Paz-Ares)
| | - Fernando Lopez-Rios
- the Department of Pathology, 12 de Octubre University Hospital, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain (Conde, Lopez-Rios)
| |
Collapse
|
7
|
Asleh K, Ouellette RJ. Tumor Copy Number Alteration Burden as a Predictor for Resistance to Immune Checkpoint Blockade across Different Cancer Types. Cancers (Basel) 2024; 16:732. [PMID: 38398121 PMCID: PMC10886982 DOI: 10.3390/cancers16040732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Immune checkpoint blockade (ICB) benefits only a subset of advanced cancer patients, and predictive biomarkers for immunotherapy response are needed. Recently, copy number alteration (CNA) burden has been proposed to predict ICB resistance. We assessed this finding using the publicly accessible data for 1661 ICB-treated patients whose tumors were profiled by MSK-IMPACT, an approved targeted assay in clinical care. We tested the hypothesis that the continuous increase in CNA burden is associated with poor overall survival following ICB. In addition, we hypothesized that the combinatorial biomarkers of tumor mutational burden (TMB) and CNA burden would better stratify patients for immune status and ICB response. Of the 1661 cases, 79% (n = 1307) were treated with anti PD-1/PD-L1 and the remaining 21% (n = 354) with anti CTLA-4 or the combination of both. In a multivariate analysis, increase in CNA burden was associated with poor overall survival [HR = 1.52, 95% CI (1.01-2.30), p = 0.04]. The combination of biomarkers TMB and CNA burden stratified patients into four clinically distinct subsets among which "LowTMB/HighCNA" showed the worst survival (p < 0.0001). The four patient subsets had unique CNA profiles and enriched pathways, which could predict transcriptional and phenotypic effects related to immune signaling and CD8+ T-cell abundance in the tumor microenvironment. CNA burden was associated with poor overall survival in patients receiving ICB and could improve patient stratification when incorporated with TMB. These findings may guide patient selection for immunotherapy or alternative strategies.
Collapse
Affiliation(s)
- Karama Asleh
- Department of Pathology and Laboratory Medicine, Halifax, NS B3H 1V8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 0A2, Canada;
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Rodney J. Ouellette
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 0A2, Canada;
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Dr. Georges L. Dumont University Hospital, Vitalité Health Network, Moncton, NB E1C 2Z3, Canada
| |
Collapse
|
8
|
Ishimaru S, Shimoi T, Sunami K, Nakajima M, Ando Y, Okita N, Nakamura K, Shibata T, Fujiwara Y, Yamamoto N. Platform trial for off-label oncology drugs using comprehensive genomic profiling under the universal public healthcare system: the BELIEVE trial. Int J Clin Oncol 2024; 29:89-95. [PMID: 38112833 PMCID: PMC10808137 DOI: 10.1007/s10147-023-02439-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/08/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Precision medicine has transformed cancer treatment by focusing on personalized approaches based on genomic abnormalities. However, comprehensive genomic profiling (CGP) and access to targeted therapies are limited in Japan. This study investigates the BELIEVE trial, which aims to improve drug accessibility for patients with actionable genetic abnormalities through off-label drug administration. METHODS The BELIEVE trial is a platform trial with a single master protocol, conducted under the Clinical Trials Act and the patient-proposed health services (PPHS) scheme. Eligible patients with solid tumors exhibiting actionable alterations were enrolled, and CGP tests covered by national health insurance were employed. Treatment selection, study drugs from collaborating pharmaceutical companies, and treatment schedules adhered to predefined protocols. Primary and secondary endpoints were evaluated, and statistical analysis was conducted based on patient response rates. RESULTS The BELIEVE trial offered treatment opportunities for patients with relapse/refractory disease who lacked standard therapies or clinical trial options. This study addresses unmet medical needs and contributes to the establishment of precision medicine systems. Similar trials like NCI-MATCH and TAPUR are being conducted globally. The BELIEVE trial provides a platform for off-label drug administration, collects essential clinical data, and contributes to drug approval applications. CONCLUSION The BELIEVE trial provides hope for patients with actionable genetic abnormalities by facilitating access to targeted therapies through off-label drug administration. It establishes a regulatory framework and promotes collaboration between industry and academia by expanding organ-specific and cross-organ biomarker-based treatments.
Collapse
Affiliation(s)
- Sae Ishimaru
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Miho Nakajima
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yayoi Ando
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Natsuko Okita
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Kenichi Nakamura
- Department of International Clinical Development/Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Taro Shibata
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
9
|
Xiang S, Lu X. Selective type II TRK inhibitors overcome xDFG mutation mediated acquired resistance to the second-generation inhibitors selitrectinib and repotrectinib. Acta Pharm Sin B 2024; 14:517-532. [PMID: 38322338 PMCID: PMC10840435 DOI: 10.1016/j.apsb.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/15/2023] [Accepted: 10/24/2023] [Indexed: 02/08/2024] Open
Abstract
Neurotrophic receptor kinase (NTRK) fusions are actionable oncogenic drivers of multiple pediatric and adult solid tumors, and tropomyosin receptor kinase (TRK) has been considered as an attractive therapeutic target for "pan-cancer" harboring these fusions. Currently, two generations TRK inhibitors have been developed. The representative second-generation inhibitors selitrectinib and repotrectinib were designed to overcome clinic acquired resistance of the first-generation inhibitors larotrectinib or entrectinib resulted from solvent-front and gatekeeper on-target mutations. However, xDFG (TRKAG667C/A/S, homologous TRKCG696C/A/S) and some double mutations still confer resistance to selitrectinib and repotrectinib, and overcoming these resistances represents a major unmet clinical need. In this review, we summarize the acquired resistance mechanism of the first- and second-generation TRK inhibitors, and firstly put forward the emerging selective type II TRK inhibitors to overcome xDFG mutations mediated resistance. Additionally, we concluded our perspectives on new challenges and future directions in this field.
Collapse
Affiliation(s)
- Shuang Xiang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| |
Collapse
|
10
|
Ziogas DC, Papadopoulou E, Gogas H, Sakellariou S, Felekouras E, Theocharopoulos C, Stefanou DT, Theochari M, Boukovinas I, Matthaios D, Koumarianou A, Zairi E, Liontos M, Koutsoukos K, Metaxa-Mariatou V, Kapetsis G, Meintani A, Tsaousis GN, Nasioulas G. Digging into the NGS Information from a Large-Scale South European Population with Metastatic/Unresectable Pancreatic Ductal Adenocarcinoma: A Real-World Genomic Depiction. Cancers (Basel) 2023; 16:2. [PMID: 38201431 PMCID: PMC10778112 DOI: 10.3390/cancers16010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
Despite ongoing oncological advances, pancreatic ductal adenocarcinoma (PDAC) continues to have an extremely poor prognosis with limited targeted and immunotherapeutic options. Its genomic background has not been fully characterized yet in large-scale populations all over the world. Methods: Replicating a recent study from China, we collected tissue samples from consecutive Greek patients with pathologically-confirmed metastatic/unresectable PDAC and retrospectively investigated their genomic landscape using next generation sequencing (NGS). Findings: From a cohort of 409 patients, NGS analysis was successfully achieved in 400 cases (56.50% males, median age: 61.8 years). Consistent with a previous study, KRAS was the most frequently mutated gene in 81.50% of tested samples, followed by TP53 (50.75%), CDKN2 (8%), and SMAD4 (7.50%). BRCA1/2 variants with on-label indications were detected in 2%, and 87.50% carried a variant associated with off-label treatment (KRAS, ERBB2, STK11, or HRR-genes), while 3.5% of the alterations had unknown/preliminary-studied actionability (TP53/CDKN2A). Most of HRR-alterations were in intermediate- and low-risk genes (CHEK2, RAD50, RAD51, ATM, FANCA, FANCL, FANCC, BAP1), with controversial actionability: 8% harbored a somatic non-BRCA1/2 alteration, 6 cases had a high-risk alteration (PALB2, RAD51C), and one co-presented a PALB2/BRCA2 alteration. Elevated LOH was associated with HRR-mutated status and TP53 mutations while lowered LOH was associated with KRAS alterations. Including TMB/MSI data, the potential benefit from an NGS-oriented treatment was increased from 1.91% to 13.74% (high-MSI: 0.3%, TMB > 10 muts/MB: 12.78%). TMB was slightly increased in females (4.75 vs. 4.46 muts/MB) and in individuals with age > 60 (4.77 vs. 4.40 muts/MB). About 28.41% showed PD-L1 > 1% either in tumor or immune cells, 15.75% expressed PD-L1 ≥ 10%, and only 1.18% had PD-L1 ≥ 50%. This is the largest depiction of real-world genomic characteristics of European patients with PDAC, which offers some useful clinical and research insights.
Collapse
Affiliation(s)
- Dimitrios C. Ziogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Eirini Papadopoulou
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - Helen Gogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Stratigoula Sakellariou
- First Department of Pathology, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Evangellos Felekouras
- First Department of Surgery, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Charalampos Theocharopoulos
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Dimitra T. Stefanou
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Maria Theochari
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Ioannis Boukovinas
- Department of Medical Oncology, Bioclinic Hospital, 54622 Thessaloniki, Greece;
| | | | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Eleni Zairi
- Oncology Department, St. Lukes Hospital, 55236 Thessaloniki, Greece;
| | - Michalis Liontos
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (M.L.); (K.K.)
| | - Konstantinos Koutsoukos
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (M.L.); (K.K.)
| | - Vasiliki Metaxa-Mariatou
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - George Kapetsis
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - Angeliki Meintani
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - Georgios N. Tsaousis
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - George Nasioulas
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| |
Collapse
|
11
|
Skórzewska M, Gęca K, Polkowski WP. A Clinical Viewpoint on the Use of Targeted Therapy in Advanced Gastric Cancer. Cancers (Basel) 2023; 15:5490. [PMID: 38001751 PMCID: PMC10670421 DOI: 10.3390/cancers15225490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
The development of therapies for advanced gastric cancer (GC) has made significant progress over the past few years. The identification of new molecules and molecular targets is expanding our understanding of the disease's intricate nature. The end of the classical oncology era, which relied on well-studied chemotherapeutic agents, is giving rise to novel and unexplored challenges, which will cause a significant transformation of the current oncological knowledge in the next few years. The integration of established clinically effective regimens in additional studies will be crucial in managing these innovative aspects of GC. This study aims to present an in-depth and comprehensive review of the clinical advancements in targeted therapy and immunotherapy for advanced GC.
Collapse
|
12
|
Ottaiano A, Sabbatino F, Perri F, Cascella M, Santorsola M, Nasti G. Back to the Bench: Unveiling the Tissue Context is Crucial to Enhance the Efficacy of Agnostic Therapies in Oncology. Target Oncol 2023; 18:819-820. [PMID: 37864664 DOI: 10.1007/s11523-023-01004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/23/2023]
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy.
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081, Baronissi, Salerno, Italy
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| |
Collapse
|
13
|
Zeverijn LJ, Looze EJ, Thavaneswaran S, van Berge Henegouwen JM, Simes RJ, Hoes LR, Sjoquist KM, van der Wijngaart H, Sebastian L, Geurts BS, Lee CK, de Wit GF, Espinoza D, Roepman P, Lin FP, Jansen AML, de Leng WWJ, van der Noort V, Leek LVM, de Vos FYFL, van Herpen CML, Gelderblom H, Verheul HMW, Thomas DM, Voest EE. Limited clinical activity of palbociclib and ribociclib monotherapy in advanced cancers with cyclin D-CDK4/6 pathway alterations in the Dutch DRUP and Australian MoST trials. Int J Cancer 2023; 153:1413-1422. [PMID: 37424386 DOI: 10.1002/ijc.34649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/04/2023] [Accepted: 05/23/2023] [Indexed: 07/11/2023]
Abstract
The Dutch Drug Rediscovery Protocol (DRUP) and the Australian Cancer Molecular Screening and Therapeutic (MoST) Program are similar nonrandomized, multidrug, pan-cancer trial platforms that aim to identify signals of clinical activity of molecularly matched targeted therapies or immunotherapies outside their approved indications. Here, we report results for advanced or metastatic cancer patients with tumors harboring cyclin D-CDK4/6 pathway alterations treated with CDK4/6 inhibitors palbociclib or ribociclib. We included adult patients that had therapy-refractory solid malignancies with the following alterations: amplifications of CDK4, CDK6, CCND1, CCND2 or CCND3, or complete loss of CDKN2A or SMARCA4. Within MoST, all patients were treated with palbociclib, whereas in DRUP, palbociclib and ribociclib were assigned to different cohorts (defined by tumor type and alteration). The primary endpoint for this combined analysis was clinical benefit, defined as confirmed objective response or stable disease ≥16 weeks. We treated 139 patients with a broad variety of tumor types; 116 with palbociclib and 23 with ribociclib. In 112 evaluable patients, the objective response rate was 0% and clinical benefit rate at 16 weeks was 15%. Median progression-free survival was 4 months (95% CI: 3-5 months), and median overall survival 5 months (95% CI: 4-6 months). In conclusion, only limited clinical activity of palbociclib and ribociclib monotherapy in patients with pretreated cancers harboring cyclin D-CDK4/6 pathway alterations was observed. Our findings indicate that monotherapy use of palbociclib or ribociclib is not recommended and that merging data of two similar precision oncology trials is feasible.
Collapse
Affiliation(s)
- Laurien J Zeverijn
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eleonora J Looze
- Division of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Subotheni Thavaneswaran
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - J Maxime van Berge Henegouwen
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert J Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Louisa R Hoes
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Katrin M Sjoquist
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Hanneke van der Wijngaart
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Lucille Sebastian
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Birgit S Geurts
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Chee K Lee
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Gijsbrecht F de Wit
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - David Espinoza
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Paul Roepman
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | - Frank P Lin
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Anne M L Jansen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wendy W J de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Lindsay V M Leek
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Filip Y F L de Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - David M Thomas
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Emile E Voest
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| |
Collapse
|
14
|
Saito R, Kuroda T, Yoshida H, Sudo K, Saito M, Tanabe H, Takano H, Yamada K, Kiyokawa T, Yonemori K, Kato T, Okamoto A, Kohno T. Genetic characteristics of platinum-sensitive ovarian clear cell carcinoma. Jpn J Clin Oncol 2023; 53:781-790. [PMID: 37248674 DOI: 10.1093/jjco/hyad045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
OBJECTIVE Most ovarian clear cell carcinomas are resistant to platinum-based chemotherapy, while a small subset shows a positive response. The aim of this study was to clarify the clinical, pathological and genetic characteristics of platinum-sensitive ovarian clear cell carcinomas. METHODS The study included 53 patients with stage III-IV ovarian clear cell carcinoma who had residual tumours after primary surgery and received platinum-based therapy between 2009 and 2018. A retrospective examination of platinum sensitivity was performed using the criterion of ≥6 months from the last day of first-line platinum therapy until recurrence/progression. Cases determined to be platinum-sensitive were subjected to immunohistochemical staining, genomic analyses using target sequencing (i.e. NCC Oncopanel) and homologous recombination deficiency (myChoice® HRD Plus) assays. RESULTS Of the 53 stage III-IV ovarian clear cell carcinoma cases, 11 (21%) were platinum-sensitive. These cases showed better progression-free and overall survival than platinum-resistant cases (hazard ratio = 0.16, P < 0.001). Among the seven sensitive cases whose tumour tissues were available for molecular profiling, five were pure ovarian clear cell carcinoma based on pathological and genetic features, whereas the remaining two cases were re-diagnosed as high-grade serous ovarian carcinoma. The pure ovarian clear cell carcinomas lacked BRCA1 and BRCA2 mutations, consistent with the absence of the homologous recombination deficiency phenotype, whereas two cases (40%) had ATM mutations. By contrast, the two high-grade serous ovarian carcinoma cases had BRCA1 or BRCA2 mutations associated with the homologous recombination deficiency phenotype. CONCLUSION The subset of platinum-sensitive ovarian clear cell carcinomas includes a majority with pure ovarian clear cell carcinoma features that lack the homologous recombination deficiency phenotype.
Collapse
Affiliation(s)
- Ryosuke Saito
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Takafumi Kuroda
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuki Sudo
- Department of Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Motoaki Saito
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Hiroshi Tanabe
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Department of Gynecology, National Cancer Center Hospital East, Kashiwa-shi, Chiba, Japan
| | - Hirokuni Takano
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Kyosuke Yamada
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Takako Kiyokawa
- Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
- Molecular Oncology, The Jikei University Graduate School of Medicine, Minato-ku, Tokyo, Japan
| |
Collapse
|
15
|
Farid-Kapadia M, Barton M, Bider-Canfield Z, Cheema PK, Gyawali B, Nightingale NM, Latifovic L, Conter HJ. Comparison of tumor-agnostic and tumor-specific clinical oncology trial designs: a systematic review and meta-analysis. Future Oncol 2023; 19:1741-1752. [PMID: 37283038 DOI: 10.2217/fon-2022-0974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Aim: To examine whether tumor-specific and tumor-agnostic oncology trials produce comparable estimates of objective response rate (ORR) in BRAF-altered cancers. Materials & methods: Electronic database searches were performed to identify phase I-III clinical trials testing tyrosine kinase inhibitors from 2000 to 2021. A random-effects model was used to pool ORRs. A total of 22 cohorts from five tumor-agnostic trials and 41 cohorts from 27 tumor-specific trials had published ORRs. Results: There was no significant difference between pooled ORRs from either trial design for multitumor analyses (37 vs 50%; p = 0.05); thyroid cancer (57 vs 33%; p = 0.10); non-small-cell lung cancer (39 vs 53%; p = 0.18); or melanoma (55 vs 51%; p = 0.58). Conclusion: For BRAF-altered advanced cancers, tumor-agnostic trials do not yield substantially different results from tumor-specific trials.
Collapse
Affiliation(s)
- Mufiza Farid-Kapadia
- Hoffmann-La Roche Limited, 7070 Mississauga Road, Mississauga, ON, L5N 5M8, Canada
| | - Madelyn Barton
- Hoffmann-La Roche Limited, 7070 Mississauga Road, Mississauga, ON, L5N 5M8, Canada
| | - Zoe Bider-Canfield
- Hoffmann-La Roche Limited, 7070 Mississauga Road, Mississauga, ON, L5N 5M8, Canada
| | - Parneet K Cheema
- William Osler Health System, 2100 Bovaird Drive East, Brampton, ON, L6R 3J7, Canada
| | - Bishal Gyawali
- Queens University, 15 Arch Street, Kingston, ON, K7L 3L4, Canada
| | | | - Lidija Latifovic
- IQVIA Solutions Canada, 1875 Buckhorn Gate, Mississauga, ON, L4W 5P1, Canada
| | - Henry J Conter
- Hoffmann-La Roche Limited, 7070 Mississauga Road, Mississauga, ON, L5N 5M8, Canada
| |
Collapse
|
16
|
Lee SE, Lee MS, Bang H, Kim MY, Choi YL, Oh YL. NTRK Fusion in a Cohort of BRAF p. V600E Wild-Type Papillary Thyroid Carcinomas. Mod Pathol 2023; 36:100180. [PMID: 37003481 DOI: 10.1016/j.modpat.2023.100180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 04/03/2023]
Abstract
Owing to the availability of a potent tropomyosin receptor kinase (TRK) inhibitor, it is necessary to develop an effective strategy to identify an enriched population of NTRK fusions in papillary thyroid carcinoma (PTC) in routine diagnostic practice. The reported prevalence of NTRK fusion in a large cohort of PTC is ∼3%. We performed an analysis to refine the characteristic histologic features of PTCs harboring NTRK fusions and further validate the diagnostic utility of pan-TRK immunohistochemistry as a screening tool. In this study, 450 PTCs known to harbor no BRAF p. V600E mutations were screened by pan-TRK immunohistochemistry, and the cases with TRK expression were confirmed by RNA-based next-generation sequencing assay. Eleven NTRK fusion cases were detected (2.4%), and all PTCs were classical subtypes. NTRK1 and NTRK3 were involved in the fusion with 9 different partner genes. Most cases showed similar characteristic histologic findings. Nodular permeative border, multinodular growth with a predominantly follicular pattern, extensive lymphatic invasion, and prominent internodular and intratumoral fibrosis were the characteristic histologic features of NTRK-rearranged PTCs. The ill-defined margins in the ultrasonography findings, which could not be clearly distinguished from the adjacent nontumorous thyroid tissue, were nodular permeative margins in histologic findings. Therefore, preoperative ultrasonographic findings in nodule margins were consistent with the final histologic findings. NTRK1/3 fusion in PTCs showed an overall sensitivity of 100% (95% CI, 71.51%-100%) and specificity of 100% (95% CI, 71.51%-100%) in the 22 cases examined, as confirmed with next-generation sequencing. Our study provides an integrative report of the preoperative ultrasonographic, histologic, immunohistochemical, and molecular features of NTRK-rearranged PTCs. Based on these findings, we propose an algorithmic approach for the stepwise assessment of NTRK fusions in PTCs.
Collapse
Affiliation(s)
- Seung Eun Lee
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Mi-Sook Lee
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Heejin Bang
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Mi Young Kim
- Department of Radiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Yoon-La Choi
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea; Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Young Lyun Oh
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
17
|
Klapp V, Álvarez-Abril B, Leuzzi G, Kroemer G, Ciccia A, Galluzzi L. The DNA Damage Response and Inflammation in Cancer. Cancer Discov 2023; 13:1521-1545. [PMID: 37026695 DOI: 10.1158/2159-8290.cd-22-1220] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/27/2023] [Accepted: 02/23/2023] [Indexed: 04/08/2023]
Abstract
Genomic stability in normal cells is crucial to avoid oncogenesis. Accordingly, multiple components of the DNA damage response (DDR) operate as bona fide tumor suppressor proteins by preserving genomic stability, eliciting the demise of cells with unrepairable DNA lesions, and engaging cell-extrinsic oncosuppression via immunosurveillance. That said, DDR sig-naling can also favor tumor progression and resistance to therapy. Indeed, DDR signaling in cancer cells has been consistently linked to the inhibition of tumor-targeting immune responses. Here, we discuss the complex interactions between the DDR and inflammation in the context of oncogenesis, tumor progression, and response to therapy. SIGNIFICANCE Accumulating preclinical and clinical evidence indicates that DDR is intimately connected to the emission of immunomodulatory signals by normal and malignant cells, as part of a cell-extrinsic program to preserve organismal homeostasis. DDR-driven inflammation, however, can have diametrically opposed effects on tumor-targeting immunity. Understanding the links between the DDR and inflammation in normal and malignant cells may unlock novel immunotherapeutic paradigms to treat cancer.
Collapse
Affiliation(s)
- Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Beatriz Álvarez-Abril
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Alberto Ciccia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Sandra and Edward Meyer Cancer Center, New York, New York
- Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| |
Collapse
|
18
|
Moreno L, DuBois SG, Glade Bender J, Mauguen A, Bird N, Buenger V, Casanova M, Doz F, Fox E, Gore L, Hawkins DS, Izraeli S, Jones DT, Kearns PR, Molenaar JJ, Nysom K, Pfister S, Reaman G, Smith M, Weigel B, Vassal G, Zwaan CM, Paoletti X, Iasonos A, Pearson AD. Combination Early-Phase Trials of Anticancer Agents in Children and Adolescents. J Clin Oncol 2023; 41:3408-3422. [PMID: 37015036 PMCID: PMC10414747 DOI: 10.1200/jco.22.02430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/07/2023] [Indexed: 04/06/2023] Open
Abstract
PURPOSE There is an increasing need to evaluate innovative drugs for childhood cancer using combination strategies. Strong biological rationale and clinical experience suggest that multiple agents will be more efficacious than monotherapy for most diseases and may overcome resistance mechanisms and increase synergy. The process to evaluate these combination trials needs to maximize efficiency and should be agreed by all stakeholders. METHODS After a review of existing combination trial methodologies, regulatory requirements, and current results, a consensus among stakeholders was achieved. RESULTS Combinations of anticancer therapies should be developed on the basis of mechanism of action and robust preclinical evaluation, and may include data from adult clinical trials. The general principle for combination early-phase studies is that, when possible, clinical trials should be dose- and schedule-confirmatory rather than dose-exploratory, and every effort should be made to optimize doses early. Efficient early-phase combination trials should be seamless, including dose confirmation and randomized expansion. Dose evaluation designs for combinations depend on the extent of previous knowledge. If not previously evaluated, limited evaluation of monotherapy should be included in the same clinical trial as the combination. Randomized evaluation of a new agent plus standard therapy versus standard therapy is the most effective approach to isolate the effect and toxicity of the novel agent. Platform trials may be valuable in the evaluation of combination studies. Patient advocates and regulators should be engaged with investigators early in a proposed clinical development pathway and trial design must consider regulatory requirements. CONCLUSION An optimized, agreed approach to the design and evaluation of early-phase pediatric combination trials will accelerate drug development and benefit all stakeholders, most importantly children and adolescents with cancer.
Collapse
Affiliation(s)
- Lucas Moreno
- Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Steven G. DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | | | | | - Nick Bird
- Solving Kids' Cancer UK, London, United Kingdom
| | - Vickie Buenger
- Coalition Against Childhood Cancer (CAC2), Philadelphia, PA
| | | | - François Doz
- Université Paris Cité, Paris, France
- SIREDO Centre (Care, Innovation Research in Pediatric, Adolescent and Young Adults Oncology), Institut Curie, Paris, France
| | | | - Lia Gore
- Children's Hospital Colorado, Aurora, CO
- University of Colorado, Aurora, CO
| | | | - Shai Izraeli
- Rina Zaizov Pediatric Hematology Oncology Division, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Hematological Malignancies Centre of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David T.W. Jones
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, United Kingdom
| | - Pamela R. Kearns
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pharmaceutical Sciences Utrecht University, Utrecht, the Netherlands
| | - Jan J. Molenaar
- Division of Pediatric Neurooncology, DKFZ, KiTZ
- Righospitalet, Copenhagen, Denmark
| | - Karsten Nysom
- Clinical Trial Unit and Childhood Brain Tumors, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Pfister
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | | | | | - Gilles Vassal
- Innovative Therapies for Children with Cancer, Paris, France
- ACCELERATE, Brussels, Belgium
- Gustave Roussy Cancer Centre, Paris, France
| | - Christian Michel Zwaan
- Righospitalet, Copenhagen, Denmark
- Department of Pediatric Oncology, Hematology, Erasmus MC, Sophia Children’s Hospital, the Netherlands
| | | | | | - Andrew D.J. Pearson
- Innovative Therapies for Children with Cancer, Paris, France
- ACCELERATE, Brussels, Belgium
| |
Collapse
|
19
|
Carmagnani Pestana R, Moyers JT, Roszik J, Sen S, Hong DS, Naing A, Herzog CE, Fu S, Piha-Paul SA, Rodon J, Yap TA, Karp DD, Tsimberidou AM, Pant S, Zarzour MA, Ratan R, Ravi V, Benjamin RS, Lazar AJ, Wang WL, Daw N, Gill JB, Harrison DJ, Lewis VO, Roland CL, Patel SR, Livingston JA, Somaiah N, Ludwig JA, Conley AP, Hamerschlak N, Gorlick R, Meric-Bernstam F, Subbiah V. Impact of Biomarker-Matched Therapies on Outcomes in Patients with Sarcoma Enrolled in Early-Phase Clinical Trials (SAMBA 101). Clin Cancer Res 2023; 29:1708-1718. [PMID: 37058010 PMCID: PMC10150251 DOI: 10.1158/1078-0432.ccr-22-3629] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/30/2022] [Accepted: 02/24/2023] [Indexed: 04/15/2023]
Abstract
PURPOSE Developing new therapeutics for any of the more than 100 sarcoma subtypes presents a challenge. After progression from standard therapies, patients with sarcoma may be referred for enrollment in early-phase trials. This study aimed to investigate whether enrollment in biomarker-matched early-phase clinical trials leads to better outcomes for patients with advanced sarcoma. EXPERIMENTAL DESIGN In this retrospective analysis, investigational treatment characteristics and longitudinal survival outcomes were analyzed in patients with biopsy-confirmed sarcoma enrolled in early-phase trials at MD Anderson Cancer Center from May 2006 to July 2021. RESULTS Five hundred eighty-seven patients were included [405 soft tissue, 122 bone, 60 gastrointestinal stromal tumor (GIST); median of three prior lines of therapy]. Most common subtypes were leiomyosarcoma (17.2%), liposarcoma (14.0%), and GIST (10.2%). Molecular testing was available for 511 patients (87.1%); 221 patients (37.6%) were treated in matched trials. Overall response rate was 13.1% matched compared with 4.9% in unmatched (P < 0.001); the clinical benefit rate at 6 months was 43.9% vs. 19.9% (P < 0.001). Progression-free survival was longer for patients in matched trials (median, 5.5 vs. 2.4 months; P < 0.001), and overall survival was also superior for patients in matched trials (median, 21.5 vs. 12.3 months; P < 0.001). The benefit of enrollment in matched trials was maintained when patients with GIST were excluded from the analysis. CONCLUSIONS Enrollment in biomarker-matched early-phase trials is associated with improved outcomes in heavily pretreated patients with metastatic sarcoma. Molecular testing of tumors from patients with advanced sarcoma and enrollment in matched trials is a reasonable therapeutic strategy.
Collapse
Affiliation(s)
- Roberto Carmagnani Pestana
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Justin T. Moyers
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Medicine, Division of Hematology and Oncology, The University of California, Irvine, Orange, California
| | - Jason Roszik
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shiraj Sen
- Sarah Cannon Research Institute, Nashville, Tennessee
| | - David S. Hong
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia E. Herzog
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina A. Piha-Paul
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy A. Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel D. Karp
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shubham Pant
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria A. Zarzour
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ravin Ratan
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert S. Benjamin
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J. Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Najat Daw
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jonathan B. Gill
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Douglas J. Harrison
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Valerae O. Lewis
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christina L. Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shreyaskumar R. Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - J. Andrew Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph A. Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony P. Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Richard Gorlick
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Lanka SM, Zorko NA, Antonarakis ES, Barata PC. Metastatic Castration-Resistant Prostate Cancer, Immune Checkpoint Inhibitors, and Beyond. Curr Oncol 2023; 30:4246-4256. [PMID: 37185436 PMCID: PMC10137248 DOI: 10.3390/curroncol30040323] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
The therapeutic landscape of several genitourinary malignancies has been revolutionized by the development of immune checkpoint inhibitors (ICIs); however, the utility of immunotherapies in prostate cancer has been limited, partly due to the immunologically "cold" tumor terrain of prostate cancer. As of today, pembrolizumab is the only immune checkpoint inhibitor approved for the treatment of metastatic castration resistant prostate cancer (mCRPC) in a select group of patients with high microsatellite instability (MSI-H), deficient mismatch repair (dMMR), or high tumor mutational burden (TMB). Looking ahead, several combinatorial approaches with ICIs involving radioligands, radiotherapy, PARP inhibitors, interleukin inhibitors, and cancer vaccines are exploring a potential synergistic effect. Furthermore, B7-H3 is an alternative checkpoint that may hold promise in adding to the treatment landscape of mCRPC. This review aims to summarize previous monotherapy and combination therapy trials of ICIs as well as novel immunotherapy combination therapeutic strategies and treatment targets in mCRPC.
Collapse
Affiliation(s)
- Sree M Lanka
- Deming Department of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Nicholas A Zorko
- Department of Hematology and Oncology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emmanuel S Antonarakis
- Department of Hematology and Oncology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Pedro C Barata
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Baek HS, Ha J, Ha S, Bae JS, Jung CK, Lim DJ. Initial Experiences of Selective RET Inhibitor Selpercatinib in Adults with Metastatic Differentiated Thyroid Carcinoma and Medullary Thyroid Carcinoma: Real-World Case Series in Korea. Curr Oncol 2023; 30:3020-3031. [PMID: 36975442 PMCID: PMC10046919 DOI: 10.3390/curroncol30030229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Recently, selpercatinib, a highly selective inhibitor of RET receptor tyrosine kinase, has been used for RET-altered thyroid cancer. We present four cases of patients with advanced thyroid cancer who were treated with selpercatinib. The first patient was a 63-year-old male with advanced medullary thyroid cancer (MTC) treated with vandetanib. Six months ago, he had an intracranial hemorrhage and swallowing difficulty. He started selpercatinib with percutaneous endoscopic gastrostomy (PEG). For 11 months, a partial response (PR) was observed stably with PEG administration without any more cardiovascular events. The second patient was a 67-year-old female with advanced MTC treated with vandetatib. After selpercatinib treatment, a PR was observed for most metastatic sites, including choroidal metastasis. The third patient was a 32-year-old female with advanced papillary thyroid cancer (PTC) without history of systematic treatment. For six months, a PR was observed at her metastatic site with manageable adverse events. The last patient was a 59-year-old female with advanced PTC treated with lenvatinib. She suffered from a panic disorder and pleural pain due to metastasis during lenvatinib treatment. After selpercatinib treatment, her pain and panic symptoms were improved. Facing varying clinical obstacles of the real world, selpercatinib safely proved remarkable therapeutic efficacy regardless of previous treatment or metastatic site.
Collapse
Affiliation(s)
- Han-Sang Baek
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Jeonghoon Ha
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Ja Seong Bae
- Department of Surgery, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Dong-Jun Lim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- Correspondence:
| |
Collapse
|
22
|
Christopoulos P, Schlenk R, Kazdal D, Blasi M, Lennerz J, Shah R, Budczies J, Malek N, Fröhling S, Rosenquist R, Schirmacher P, Bozorgmehr F, Kuon J, Reck M, Thomas M, Stenzinger A. Real-world data for precision cancer medicine-A European perspective. Genes Chromosomes Cancer 2023. [PMID: 36852573 DOI: 10.1002/gcc.23135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023] Open
Abstract
Leveraging real-world data (RWD) for drug access is necessary to overcome a key challenge of modern precision oncology: tackling numerous low-prevalence oncogenic mutations across cancers. Withholding a potentially active medication in patients with rare mutations for the sake of control chemotherapy or "best" supportive care is neither practicable nor ethically justifiable anymore, particularly as RWD could meanwhile be used instead, according to scientific principles outlined by the US Food and Drug Administration, European Medicines Agency and other stakeholders. However, practical implementation varies, with occasionally opposite recommendations based on the same evidence in different countries. In the face of growing need for precision drugs, more transparency of evaluation, a priori availability of guidance for the academia and industry, as well as a harmonized framework for health technology assessment across the European Union (EU) are imperative. These could in turn trigger infrastructural changes in national and pan-European registries, cancer management guidelines (e.g., frequency of routine radiologic restaging, inclusion of patient-reported outcomes), and the health data space, to ensure conformity with declared standards and facilitate extraction of RWD sets (including patient-level data) suitable for approval and pricing with minimal effort. For an EU-wide unification of precision cancer medicine, collective negotiation of drug supply contracts and funding solidarity would additionally be required to handle the financial burden. According to experience from pivotal European programs, off-label use could potentially also be harmonized across EU-states to accelerate availability of novel drugs, streamline collection of valuable RWD, and mitigate related costs through wider partnerships with pharmaceutical companies.
Collapse
Affiliation(s)
- Petros Christopoulos
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany.,Centers for Personalized Medicine (ZPM), Germany
| | - Richard Schlenk
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,NCT Trial Center, National Center of Tumor Diseases, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Kazdal
- German Center for Lung Research (DZL), Gießen, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Miriam Blasi
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Jochen Lennerz
- Machachussets General Hospital, Harvard University, Boston, USA
| | - Rajiv Shah
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Jan Budczies
- Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nisar Malek
- Centers for Personalized Medicine (ZPM), Germany.,Department of Gastroenterology, Tübingen University Hospital, Tübingen, Germany
| | - Stefan Fröhling
- Centers for Personalized Medicine (ZPM), Germany.,Department of Translational Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Peter Schirmacher
- Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| | - Farastuk Bozorgmehr
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Jonas Kuon
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany.,Department of Oncology, Lungenklinik Löwenstein, Löwenstein, Germany
| | - Martin Reck
- German Center for Lung Research (DZL), Gießen, Germany.,Department of Thoracic Oncology, Lungenclinic Großhansdorf, Großhansdorf, Germany
| | - Michael Thomas
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Albrecht Stenzinger
- German Center for Lung Research (DZL), Gießen, Germany.,Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| |
Collapse
|
23
|
Pestana RC, Serrano C. Analysis of histology-agnostic targets among soft tissue and bone sarcomas in the AACR GENIE database. Front Oncol 2023; 12:1079909. [PMID: 36741731 PMCID: PMC9890057 DOI: 10.3389/fonc.2022.1079909] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
Background The development of novel therapies for patients with sarcoma is challenging due to the rarity and diversity of these mesenchymal neoplasms. Hence, histology-agnostic approvals can be of particular interest for the treatment of patients with soft tissue and bone sarcoma. Methods We queried the American Association for Cancer Research (AACR) Project Genomics Evidence Neoplasia Information Exchange (GENIE) database Cohort v12.0-Public to investigate the prevalence of currently Food and Drug Administration (FDA)-approved and other potentially actionable histology-agnostic alterations in patients with soft tissue and bone sarcoma. Targets were identified by a literature review by the authors. Results are presented for each cohort identified in the GENIE database, namely: (1) soft tissue sarcoma (STS), (2) gastrointestinal stromal tumor (GIST), (3) bone sarcoma, (4) uterine sarcoma, and (5) breast sarcoma. Results We identified 7,512 samples of 6,955 patients with sarcoma in the AAACR GENIE database v12.0-Public. Molecular alterations that could lead to the clinical use of a currently approved histology-agnostic therapy were identified in 2.1% of sarcomas (2.6% STS, 1.3% GIST, 1.4% bone, 2.7% uterine, and 0% breast). In addition, 2.9% of patients could be eligible for future histology-agnostic approvals. These specific mutations, fusions, and amplifications occurred in multiple histotypes in all cohorts. Discussion Exploring a public large-scale genomic database, we identified that 5% of patients with sarcoma could be eligible for current histology-agnostic FDA-approved drugs or future potential histology-agnostic indications. These actionable alterations were present in a wide variety of histologies in soft tissue and bone sarcomas, highlighting that next-generation sequencing can be considered for patients with advanced sarcoma to guide treatment strategies.
Collapse
Affiliation(s)
| | - César Serrano
- Department of Medical Oncology, Vall d’Hebron University Hospital, Barcelona, Spain,Sarcoma Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), Hospital Universitario Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| |
Collapse
|
24
|
Moyers JT, Pestana RC, Roszik J, Hong DS, Naing A, Fu S, Piha-Paul S, Yap TA, Karp D, Rodon J, Livingston A, Zarzour MA, Ravi V, Patel S, Benjamin RS, Ludwig J, Herzog C, Ratan R, Somaiah N, Conley A, Gorlick R, Meric-Bernstam F, Subbiah V. Examining Stripes on a Herd of Zebras: Impact of Genomic Matching for Ultrarare Sarcomas in Phase 1 Clinical Trials (SAMBA 102). Clin Cancer Res 2023; 29:401-409. [PMID: 36288393 PMCID: PMC9843435 DOI: 10.1158/1078-0432.ccr-22-2509] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/04/2022] [Accepted: 10/24/2022] [Indexed: 01/21/2023]
Abstract
PURPOSE Recently, the Connective Tissue Oncology Society published consensus guidelines for recognizing ultrarare sarcomas (URS), defined as sarcomas with an incidence ≤1 per 1,000,000. We assessed the outcomes of 56 patients with soft tissue, and 21 with bone sarcomas, enrolled in Phase 1 trials. EXPERIMENTAL DESIGN In this Sarcoma-Matched Biomarker Analysis (SAMBA-102 study), we reviewed records from patients on Phase 1 trials at the University of Texas MD Anderson Cancer Center between January 2013 and June 2021. RESULTS Among 587 sarcomas, 106 (18.1%) were classified as URS. Fifty (47%) were male, and the median age was 44.3 years (range, 19-82). The most common subtypes were alveolar soft part sarcoma (ASPS), chordoma, dedifferentiated chondrosarcoma, and sclerosing epithelioid fibrosarcoma. Compared with common sarcomas, median OS was similar 16.1 months [95% confidence interval (CI), 13.6-17.5] versus 16.1 (95% CI, 8.2-24.0) in URS (P = 0.359). Objective response to treatment was higher in URS 13.2% (n = 14/106) compared with common sarcomas 6.9% (n = 33/481; P = 0.029). Median OS for those treated on matched trials was 27.3 months (95% CI, 1.9-52.7) compared with 13.4 months (95% CI, 6.3-20.6) for those not treated on matched trials (P = 0.291). Eight of 33 (24%) molecularly matched treatments resulted in an objective response, whereas 6 of 73 unmatched treatments (8.2%) resulted in an objective response (P = 0.024). Clinical benefit rate was 36.4% (12/33) in matched trials versus 26.0% (19/73) in unmatched trials (P = 0.279). CONCLUSIONS The results demonstrate the benefit of genomic selection in Phase 1 trials to help identify molecular subsets likely to benefit from targeted therapy.
Collapse
Affiliation(s)
- Justin T. Moyers
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Division of Hematology and Oncology, Department of Medicine, University of California, Irvine, Orange, California
| | - Roberto Carmagnani Pestana
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jason Roszik
- Division of Cancer Medicine, Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S. Hong
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina Piha-Paul
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy A. Yap
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Karp
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jordi Rodon
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andy Livingston
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Alejandra Zarzour
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vinod Ravi
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shreyaskumar Patel
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert S. Benjamin
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph Ludwig
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia Herzog
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ravin Ratan
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeta Somaiah
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony Conley
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard Gorlick
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Corresponding Author: Vivek Subbiah, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 455, PO Box 301402, Houston, TX 77030. E-mail:
| |
Collapse
|
25
|
Vingiani A, Lorenzini D, Conca E, Volpi CC, Trupia DV, Gloghini A, Perrone F, Tamborini E, Dagrada GP, Agnelli L, Capone I, Busico A, Pruneri G. Pan-TRK immunohistochemistry as screening tool for NTRK fusions: A diagnostic workflow for the identification of positive patients in clinical practice. Cancer Biomark 2023; 38:301-309. [PMID: 37545217 DOI: 10.3233/cbm-220357] [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] [Indexed: 08/08/2023]
Abstract
BACKGROUND Pan-TRK inhibitors Entrectinib and Larotrectinib have been recently approved as tumor-agnostic therapies in NTRK1-2-3 rearranged patients and there is therefore an urgent need to identify reliable and accessible biomarkers for capturing NTRK fusions in the real-world practice. OBJECTIVE We aim to assess the analytical validity of the recently released pan-TRK assay (Ventana), running a head-to-head comparison between immunohistochemistry and Archer FusionPlex Lung Panel (ArcherDX) that is designed to detect key fusions in 13 genes, also including NTRK1-3. METHODS Pan-TRK IHC and NGS analysis were conducted on a retrospective/prospective cohort of 124 cancer patients (carcinomas, 93 cases; soft tissue sarcomas, 19; primary central nervous system tumours, 10; and neuroblastomas, 2). FISH data were available in most of the IHC/NGS discordant cases. RESULTS A comparison between IHC and NGS results was carried out in 117 cases: among 30 pan-TRK positive cases, NTRK rearrangement by NGS was found in 11 (37%), while one of the 87 (1.1%) pan-TRK negative cases (a case of NSCLC) showed a TPM3-NRTK1 rearrangement by NGS. Accordingly, sensitivity and specificity of IHC in predicting NTRK status were 91.7% and 81.9%, respectively, while negative (NPV) and positive predictive value (PPV) were 98.8% and 36.7%, respectively. CONCLUSIONS These data lead to suggest that IHC with VENTANA pan-TRK antibody can be a reliable screening tool for the identification of patients potentially bearing NTRK rearranged tumours.
Collapse
Affiliation(s)
- Andrea Vingiani
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Daniele Lorenzini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Elena Conca
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Chiara Costanza Volpi
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Desirè Viola Trupia
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Federica Perrone
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Elena Tamborini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Gian Paolo Dagrada
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Luca Agnelli
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Iolanda Capone
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Adele Busico
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
| | - Giancarlo Pruneri
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
| |
Collapse
|
26
|
Jiao XD, Qin BD, Wang Z, Liu K, Wu Y, Ling Y, Qin WX, Wang MM, Yuan LY, Barreto SG, Kim AW, Mak K, Li H, Xu YY, Qiu XM, Wu M, Jin M, Xu LC, Zhong Y, Yang H, Chen XQ, Zeng Y, Shi J, Zhu WY, Ding QQ, Jia W, Liu SF, Zhou JJ, Shen H, Yao SH, Guo ZJ, Li T, Zhou PJ, Dong XW, Lu WF, Coleman RL, Akce M, Akladios C, Puccetti F, Zang YS. Targeted therapy for intractable cancer on the basis of molecular profiles: An open-label, phase II basket trial (Long March Pathway). Front Oncol 2023; 13:860711. [PMID: 36910668 PMCID: PMC9995917 DOI: 10.3389/fonc.2023.860711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Purpose We evaluated he effects of molecular guided-targeted therapy for intractable cancer. Also, the epidemiology of druggable gene alterations in Chinese population was investigated. Materials and methods The Long March Pathway (ClinicalTrials.gov identifier: NCT03239015) is a non-randomized, open-label, phase II trial consisting of several basket studies examining the molecular profiles of intractable cancers in the Chinese population. The trial aimed to 1) evaluate the efficacy of targeted therapy for intractable cancer and 2) identify the molecular epidemiology of the tier II gene alterations among Chinese pan-cancer patients. Results In the first stage, molecular profiles of 520 intractable pan-cancer patients were identified, and 115 patients were identified to have tier II gene alterations. Then, 27 of these 115 patients received targeted therapy based on molecular profiles. The overall response rate (ORR) was 29.6% (8/27), and the disease control rate (DCR) was 44.4% (12/27). The median duration of response (DOR) was 4.80 months (95% CI, 3.33-27.2), and median progression-free survival (PFS) was 4.67 months (95% CI, 2.33-9.50). In the second stage, molecular epidemiology of 17,841 Chinese pan-cancer patients demonstrated that the frequency of tier II gene alterations across cancer types is 17.7%. Bladder cancer had the most tier-II alterations (26.1%), followed by breast cancer (22.4%), and non-small cell lung cancer (NSCLC; 20.2%). Conclusion The Long March Pathway trial demonstrated a significant clinical benefit for intractable cancer from molecular-guided targeted therapy in the Chinese population. The frequency of tier II gene alterations across cancer types supports the feasibility of molecular-guided targeted therapy under basket trials.
Collapse
Affiliation(s)
- Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ying Wu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yan Ling
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wen-Xing Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Miao-Miao Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ling-Yan Yuan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | | | - Anthony W Kim
- Division of Thoracic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Kimberley Mak
- Department of Radiation Oncology, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| | - Hao Li
- Department of Medical Oncology, Shanghai Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yuan-Yuan Xu
- Department of Surgical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Ming Qiu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Min Wu
- Department Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Chao Xu
- Department of Interventional Radiology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yi Zhong
- Department of Medical Oncology, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Yang
- Department of Medical Oncology, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Xue-Qin Chen
- Department of Medical Oncology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Zeng
- Department of Pathology, Shanghai Tongji Hospital, Shanghai Tongji University, Shanghai, China
| | - Jun Shi
- Department of Gastrointestinal Surgery, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Wen-Yu Zhu
- Department of Medical Oncology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Qing-Qing Ding
- Department of Geriatric Oncology, Jiangsu Provincial People's Hospital, Nanjing Medical University, Nanjing, China
| | - Wei Jia
- Department of Respiratory, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Su-Fen Liu
- Department of Gynecology, Changzhou No.2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Jun-Jing Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Wuxi No.4 People's Hospital, Jiangnan University, Wuxi, China
| | - Hong Shen
- Department of Medical Oncology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shi-Hua Yao
- Department of Thoracic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhao-Ji Guo
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ting Li
- Department of Medical Oncology, Shanghai Cancer Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Pei-Juan Zhou
- Department of Traditional Chinese Medicine, Shanghai Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Xue-Wei Dong
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, Soochow University, Changzhou, China
| | - Wen-Feng Lu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mehmet Akce
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Chérif Akladios
- Department of Obstetrics and Gynecology, University of Strasbourg, Strasbourg, France
| | - Francesco Puccetti
- Department of Gastrointestinal Surgery, San Raffaele Hospital IRCCS, Milan, Italy
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
27
|
A scoping review and meta-analysis on the prevalence of pan-tumour biomarkers (dMMR, MSI, high TMB) in different solid tumours. Sci Rep 2022; 12:20495. [PMID: 36443366 PMCID: PMC9705554 DOI: 10.1038/s41598-022-23319-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 10/29/2022] [Indexed: 11/29/2022] Open
Abstract
Immune checkpoint inhibitors have been approved in the USA for tumours exhibiting mismatch repair deficiency (dMMR), microsatellite instability (MSI), or high tumour mutational burden (TMB), with regulatory and reimbursement applications in multiple other countries underway. As the estimated budget impacts of future reimbursements depend on the size of the potential target population, we performed a scoping review and meta-analysis of the prevalence of these pan-tumour biomarkers in different cancers. We systematically searched Medline/Embase and included studies reporting the prevalence of dMMR/MSI/high TMB in solid tumours published 01/01/2018-31/01/2021. Meta-analyses were performed separately for the pan-cancer prevalence of each biomarker, and by cancer type and stage where possible. The searches identified 3890 papers, with 433 prevalence estimates for 32 different cancer types from 201 studies included in meta-analyses. The pooled overall prevalence of dMMR, MSI and high TMB (≥ 10 mutations/Mb) in pan-cancer studies was 2.9%, 2.7% and 14.0%, respectively. The prevalence profiles of dMMR/MSI and high TMB differed across cancer types. For example, endometrial, colorectal, small bowel and gastric cancers showed high prevalence of both dMMR and MSI (range: 8.7-26.8% and 8.5-21.9%, respectively) and high TMB (range: 8.5-43.0%), while cervical, esophageal, bladder/urothelial, lung and skin cancers showed low prevalence of dMMR and MSI (< 5%), but high prevalence of high TMB (range: 23.7-52.6%). For other cancer types, prevalence of all three biomarkers was generally low (< 5%). This structured review of dMMR/MSI/high TMB prevalence across cancers and for specific cancer types and stages provide timely evidence to inform budget impact forecasts in health technology assessments for drug approvals based on these pan-tumour biomarkers.
Collapse
|
28
|
NTRK Gene Fusions in Solid Tumors and TRK Inhibitors: A Systematic Review of Case Reports and Case Series. J Pers Med 2022; 12:jpm12111819. [PMID: 36579526 PMCID: PMC9695027 DOI: 10.3390/jpm12111819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
The approval of larotrectinib and entrectinib for cancer patients harboring an NTRK gene fusion has represented a milestone in the era of "histology-agnostic" drugs. Among the clinical trials that led to the approval of these two drugs, most of the enrolled patients were affected by soft tissue sarcomas, lung, and salivary gland cancer. However, as next-generation sequencing assays are increasingly available in the clinical setting, health care professionals may be able to detect NTRK gene fusions in patients affected by tumor types under or not represented in the clinical trials. To this aim, we systematically reviewed MEDLINE from its inception to 31 August 2022 for case reports and case series on patients with NTRK gene fusion-positive tumors treated with TRK inhibitors. A virtual cohort of 43 patients was created, excluding those enrolled in the above-mentioned clinical trials. Although our results align with those existing in the literature, various cases of central nervous system tumors were registered in our cohort, confirming the benefit of these agents in this subgroup of patients. Large, multi-institutional registries are needed to provide more information about the efficacy of TRK inhibitors in cancer patients affected by tumor types under or not represented in the clinical trials.
Collapse
|
29
|
Normanno N, Apostolidis K, Wolf A, Al Dieri R, Deans Z, Fairley J, Maas J, Martinez A, Moch H, Nielsen S, Pilz T, Rouleau E, Patton S, Williams V. Access and quality of biomarker testing for precision oncology in Europe. Eur J Cancer 2022; 176:70-77. [PMID: 36194905 DOI: 10.1016/j.ejca.2022.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Predictive biomarkers are essential for selecting the best therapeutic strategy in patients with cancer. The International Quality Network for Pathology, the European Cancer Patient Coalition and the European Federation of Pharmaceuticals Industries and Associations evaluated the access to and quality of biomarker testing across Europe. METHODS Data sources included surveys of 141 laboratory managers and 1.665 patients, and 58 in-depth interviews with laboratory managers, physicians and payers. Four access metrics (laboratory access, test availability, test reimbursement, test order rate) and three quality metrics (quality scheme participation, laboratory accreditation, test turnaround time) were applied to rank the results. RESULTS The access to precision medicines is higher in countries with public national reimbursement processes in place. Lack of diagnostic laboratory infrastructure, inefficient organization and/or insufficient public reimbursement narrow the access to single biomarker tests in many European countries. In countries with limited public reimbursement, pharma and patients' out of pocket were the primary funding sources for testing. Uptake of multi-biomarker next generation sequencing (NGS) is highly varied, ranging from 0% to >50%. Financial constraints, a lack of NGS testing capabilities and the failure to include NGS testing in the guidelines represent the main barriers to NGS implementation. The quality of biomarker testing is highest in Western and Northern Europe, with more than 90% of laboratories participating in quality assurance schemes. CONCLUSIONS Our data clearly indicate the need for a call to action to ensure the clinical implementation of precision medicine in Europe.
Collapse
Affiliation(s)
- Nicola Normanno
- Istituto Nazionale Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy.
| | | | | | - Raed Al Dieri
- European Society of Pathology (ESP), Brussels, Belgium
| | | | | | - Jörg Maas
- Deutsche Gesellschaft für Pathologie E.V. (DGP), Berlin, Germany
| | | | - Holger Moch
- University Hospital and University, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
30
|
Hobbs BP, Pestana RC, Zabor EC, Kaizer AM, Hong DS. Basket Trials: Review of Current Practice and Innovations for Future Trials. J Clin Oncol 2022; 40:3520-3528. [PMID: 35537102 PMCID: PMC10476732 DOI: 10.1200/jco.21.02285] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/06/2021] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
Advances in biology and immunology have elucidated genetic and immunologic origins of cancer. Innovations in sequencing technologies revealed that distinct cancer histologies shared common genetic and immune phenotypic traits. Pharmacologic developments made it possible to target these alterations, yielding novel classes of targeted agents whose therapeutic potential span multiple tumor types. Basket trials, one type of master protocol, emerged as a tool for evaluating biomarker-targeted therapies among multiple tumor histologies. Conventionally conducted within the phase II setting and designed to estimate high and durable objective responses, basket trials pose challenges to statistical design and interpretation of results. This article reviews basket trials implemented in oncology studies and discusses issues related to their statistical design and analysis.
Collapse
Affiliation(s)
- Brian P. Hobbs
- Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Roberto Carmagnani Pestana
- Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Emily C. Zabor
- Quantitative Health Sciences & Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Alexander M. Kaizer
- Biostatistics and Informatics, University of Colorado-Anschutz Medical Campus, Aurora, CO
| | - David S. Hong
- Investigational Cancer Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
31
|
Sartore-Bianchi A, Agostara AG, Patelli G, Mauri G, Pizzutilo EG, Siena S. Application of histology-agnostic treatments in metastatic colorectal cancer. Dig Liver Dis 2022; 54:1291-1303. [PMID: 35701319 DOI: 10.1016/j.dld.2022.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023]
Abstract
Cancer treatment is increasingly focused on targeting molecular alterations identified across different tumor histologies. While some oncogenic drivers such as microsatellite instability (MSI) and NTRK fusions are actionable with the very same approach regardless of tumor type ("histology-agnostic"), others require histology-specific therapeutic adjustment ("histology-tuned") by means of adopting specific inhibitors and ad hoc combinations. Among histology-agnostic therapies, pembrolizumab or dostarlimab demonstrated comparable activity in MSI metastatic colorectal cancer (mCRC) as in other tumors with MSI status (ORR 38% vs 40% and 36% vs 39%, respectively), while entrectinib or larotrectinib proved effective in NTRK rearranged mCRC even though less dramatically than in the overall population (ORR 20% vs 57%, and 50% vs 78%, respectively). Histology-tuned approaches in mCRC are those targeting BRAFV600E mutations and ERBB2 amplification, highlighting the need of simultaneous anti-EGFR blockade or careful choice of companion inhibitors in this tumor type. Anti-RET and anti-ALK therapies emerged as a potential histology-agnostic indications, while anti-KRASG12C strategies could develop as future histology-tuned therapies. Targeting of ERBB2 mutations and NRG1 fusion provided discrepant results. In conclusion, agnostic targets such as MSI and NTRK fusions are already exploitable in mCRC, while the plethora of emerging histology-tuned targets represent a challenging opportunity requiring concurrent evolution of molecular diagnostic tools.
Collapse
Affiliation(s)
- Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Milano, Università degli Studi di Milano Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Alberto Giuseppe Agostara
- Department of Oncology and Hemato-Oncology, Milano, Università degli Studi di Milano Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Milano, Università degli Studi di Milano Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Gianluca Mauri
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy; IFOM-FIRC Institute of Molecular Oncology, Milan, Italy
| | - Elio Gregory Pizzutilo
- Department of Oncology and Hemato-Oncology, Milano, Università degli Studi di Milano Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Milano, Università degli Studi di Milano Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy.
| |
Collapse
|
32
|
Horgan D, Spanic T, Apostolidis K, Curigliano G, Chorostowska-Wynimko J, Dauben HP, Lal JA, Dziadziuszko R, Mayer-Nicolai C, Kozaric M, Jönsson B, Gutierrez-Ibarluzea I, Fandel MH, Lopert R. Towards Better Pharmaceutical Provision in Europe-Who Decides the Future? Healthcare (Basel) 2022; 10:1594. [PMID: 36011250 PMCID: PMC9408332 DOI: 10.3390/healthcare10081594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
Significant progress has been achieved in human health in the European Union in recent years. New medicines, vaccines, and treatments have been developed to tackle some of the leading causes of disease and life-threatening illnesses. It is clear that investment in research and development (R&D) for innovative medicines and treatments is essential for making progress in preventing and treating diseases. Ahead of the legislative process, which should begin by the end of 2022, discussions focus on how Europe can best promote the huge potential benefits of new science and technology within the regulatory framework. The challenges in European healthcare were spelled out by the panellists at the roundtable organised by European Alliance for Personalised Medicine (EAPM). Outcomes from panellists' discussions have been summarized and re-arranged in this paper under five headings: innovation, unmet medical need, access, security of supply, adapting to progress, and efficiency. Some of the conclusions that emerged from the panel are a call for a better overall holistic vision of the future of pharmaceuticals and health in Europe and a collaborative effort among all stakeholders, seeing the delivery of medicines as part of a broader picture of healthcare.
Collapse
Affiliation(s)
- Denis Horgan
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Faculty of Engineering and Technology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
| | - Tanja Spanic
- Europa Donna, The European Breast Cancer Coalition, 20149 Milan, Italy
| | | | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- European Institute of Oncology, IRCCS, 20139 Milan, Italy
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka Str., 01-138 Warsaw, Poland
| | | | - Jonathan A. Lal
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Faculty of Engineering and Technology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
- Institute for Public Health Genomics, Department of Genetics and Cell Biology, GROW School of Oncology and Developmental Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, P.O. Box 616, 6211 LK Maastricht, The Netherlands
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy of the Medical University of Gdansk, 80-214 Gdansk, Poland
| | | | - Marta Kozaric
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
| | - Bengt Jönsson
- Department of Economics, Stockholm School of Economics (SSE), P.O. Box 6501, 113 83 Stockholm, Sweden
| | - Iñaki Gutierrez-Ibarluzea
- Department of Knowledge Management and Evaluation of the Basque Foundation for Health Innovation and Research (BIOEF), 48902 Barakaldo, Spain
| | | | - Ruth Lopert
- Organisation for Economic Co-Operation and Development, OECD, 75775 Paris, France
| |
Collapse
|
33
|
Kaizer A, Zabor E, Nie L, Hobbs B. Bayesian and frequentist approaches to sequential monitoring for futility in oncology basket trials: A comparison of Simon's two-stage design and Bayesian predictive probability monitoring with information sharing across baskets. PLoS One 2022; 17:e0272367. [PMID: 35917296 PMCID: PMC9345361 DOI: 10.1371/journal.pone.0272367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
This article discusses and compares statistical designs of basket trial, from both frequentist and Bayesian perspectives. Baskets trials are used in oncology to study interventions that are developed to target a specific feature (often genetic alteration or immune phenotype) that is observed across multiple tissue types and/or tumor histologies. Patient heterogeneity has become pivotal to the development of non-cytotoxic treatment strategies. Treatment targets are often rare and exist among several histologies, making prospective clinical inquiry challenging for individual tumor types. More generally, basket trials are a type of master protocol often used for label expansion. Master protocol is used to refer to designs that accommodates multiple targets, multiple treatments, or both within one overarching protocol. For the purpose of making sequential decisions about treatment futility, Simon's two-stage design is often embedded within master protocols. In basket trials, this frequentist design is often applied to independent evaluations of tumor histologies and/or indications. In the tumor agnostic setting, rarer indications may fail to reach the sample size needed for even the first evaluation for futility. With recent innovations in Bayesian methods, it is possible to evaluate for futility with smaller sample sizes, even for rarer indications. Novel Bayesian methodology for a sequential basket trial design based on predictive probability is introduced. The Bayesian predictive probability designs allow interim analyses with any desired frequency, including continual assessments after each patient observed. The sequential design is compared with and without Bayesian methods for sharing information among a collection of discrete, and potentially non-exchangeable tumor types. Bayesian designs are compared with Simon's two-stage minimax design.
Collapse
Affiliation(s)
- Alexander Kaizer
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Anschutz Medical Campus, Aurora, CO, United States of America
| | - Emily Zabor
- Department of Quantitative Health Sciences & Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States of America
| | - Lei Nie
- Division of Biometrics II, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| | - Brian Hobbs
- Department of Population Health, University of Texas-Austin, Austin, TX, United States of America
| |
Collapse
|
34
|
The Role of Histology-Agnostic Drugs in the Treatment of Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2022; 23:ijms23158535. [PMID: 35955671 PMCID: PMC9369092 DOI: 10.3390/ijms23158535] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023] Open
Abstract
Precision medicine has opened up a new era in the development of anti-cancer agents that is focused on identifying biomarkers predictive of treatment response regardless of tumor histology. Since 2017, the Food and Drug Administration has approved six drugs with histology-agnostic indications: pembrolizumab (both for tumors with the mismatch-repair deficiency (dMMR)/high microsatellite instability (MSI-H) phenotype and for those with the high tumor mutational burden (TMB-H) phenotype), dostarlimab (for dMMR tumors), larotrectinib and entrectinib (for tumors harboring neurotrophic tyrosine receptor kinase (NTRK) fusions), and the combination of dabrafenib plus trametinib (for BRAF V600E-mutated tumors). The genomic alterations targeted by these antineoplastic agents are rare in metastatic castration-resistant prostate cancer (mCRPC). Furthermore, only a small number of mCRPC patients were enrolled in the clinical trials that led to the approval of the above-mentioned drugs. Therefore, we critically reviewed the literature on the efficacy of histology-agnostic drugs in mCRPC patients. Although the available evidence derives from retrospective studies and case reports, our results confirmed the efficacy of pembrolizumab in dMMR/MSI-H mCRPC. In contrast, few data are available for dostarlimab, larotrectinib, entrectinib, and dabrafenib-trametinib in this subset of patients. Large, multi-institutional registries aimed at collecting real-world data are needed to better comprehend the role of tissue-agnostic drugs in mCRPC patients.
Collapse
|
35
|
Corti C, Antonarelli G, Valenza C, Nicolò E, Rugo H, Cortés J, Harbeck N, Carey LA, Criscitiello C, Curigliano G. Histology-agnostic approvals for antibody-drug conjugates in solid tumours: is the time ripe? Eur J Cancer 2022; 171:25-42. [PMID: 35696887 DOI: 10.1016/j.ejca.2022.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/11/2022] [Accepted: 04/29/2022] [Indexed: 11/15/2022]
Abstract
Several antibody-drug conjugates (ADCs) have been recently approved to treat solid tumours. Since ADCs seem to have activity in multiple malignancies sharing the expression of a specific antigen, they may be mirroring the experience of histology-agnostic-targeted treatments. So, the possibility to interpret the activity of some ADCs across different cancer types in a biomarker-driven perspective arises. However, relevant biological, methodological, and regulatory challenges should be highlighted and addressed, in order to grant ADCs biomarker-driven regulatory approvals in the next future. In this review, we discuss challenges and opportunities posed by the pan-histological expansion of ADCs in solid tumours. In particular, we provide an overview about technological and manufacturing advancements; we offer up-to-date highlights of the current evidence from clinical trials investigating ADCs in solid tumours; we discuss the need for the identification of optimal predictive biomarkers, as well as major methodological, statistical, and regulatory considerations for a biomarker-driven histology-agnostic approach.
Collapse
Affiliation(s)
- Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Eleonora Nicolò
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Hope Rugo
- San Francisco, UCSF Helen Diller Family Comprehensive Cancer Center Precision Medicine Cancer Building, San Francisco, CA, USA
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Quironsalud Group, Barcelona, Spain; Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Spain
| | - Nadia Harbeck
- Breast Center, Dept OB&GYN and CCCMunich, LMU University Hospital, Munich, Germany
| | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
36
|
Gerratana L, Movarek M, Wehbe F, Katam N, Mahalingam D, Donahue J, Shah A, Chae YK, Mulcahy M, Tsarwhas D, Villaflor V, Kalyan A, Hussein M, Patel J, Chandra S, Platanias LC, Gradishar W, Cristofanilli M, Behdad A. Genomic Landscape of Advanced Solid Tumors in Circulating Tumor DNA and Correlation With Tissue Sequencing: A Single Institution's Experience. JCO Precis Oncol 2022; 6:e2100289. [PMID: 35772051 DOI: 10.1200/po.21.00289] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Circulating tumor DNA (ctDNA) has emerged as a promising noninvasive biomarker for baseline characterization and longitudinal monitoring of a tumor throughout disease management. The aim of this study was to evaluate the utility of ctDNA across a wide spectrum of tumor types. METHODS We retrospectively identified 1,763 patients with advanced cancers who had next-generation sequencing of ctDNA or tumor tissue completed by a designated commercial assay at Northwestern University. RESULTS ctDNA identified at least one gene alteration in 90% of patients. The number of detected alterations (NDA) and mutant allele frequency (MAF) of the most frequently mutated genes varied significantly across tumor types, with the highest MAF observed in gastric, colorectal, and breast cancers and the highest NDA observed in colorectal, lung squamous, and ovarian/endometrial cancers. TP53 was the most mutated gene in all tumor types. PIK3CA, ERBB2, BRCA1, and FGFR1 alterations were associated with breast cancer, and ESR1 mutations were exclusively detected in this tumor type. Colorectal cancer was characterized by alterations in KRAS and APC mutations, whereas KRAS, EGFR, PIK3CA, and BRAF mutations were common in lung adenocarcinoma. Concordance between blood and tissue sequencing was notably observed for truncal gene alterations (eg, APC and KRAS), whereas low concordance was often observed in genes associated with treatment resistance mechanisms (eg, RB1 and NF1). Tumor mutational burden (TMB) varied significantly across tumor types, and patients with high MAF or NDA had a significantly higher TMB score with one of the investigated platforms. CONCLUSION The study provided new insights into the ctDNA mutational landscape across solid tumors, suggesting new hypotheses-generating data and caveats for future histotype-agnostic workflows integrated with tissue-based biomarkers such as TMB.
Collapse
Affiliation(s)
- Lorenzo Gerratana
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL.,Department of Medicine, University of Udine, Udine, Italy.,Department of Medical Oncology, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Michael Movarek
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine/New York-Presbyterian Hospital, New York, NY
| | - Firas Wehbe
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Neelima Katam
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Devalingam Mahalingam
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Jeannine Donahue
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Ami Shah
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Young K Chae
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Mary Mulcahy
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Dean Tsarwhas
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Victoria Villaflor
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Aparna Kalyan
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Maha Hussein
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Jyoti Patel
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Sunandana Chandra
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Leonidas C Platanias
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - William Gradishar
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Massimo Cristofanilli
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL.,Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine/New York-Presbyterian Hospital, New York, NY
| | - Amir Behdad
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
37
|
Challenges of Systemic Therapy Investigations for Bone Sarcomas. Int J Mol Sci 2022; 23:ijms23073540. [PMID: 35408900 PMCID: PMC8998654 DOI: 10.3390/ijms23073540] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Bone sarcoma is a rare component of malignant solid tumors that accounts for only ~0.2% of malignancies. Bone sarcomas present various histological types, and genomic mutations differ markedly by the histological types. Although there are vast mutations in various bone sarcomas, most of them are non-actionable, and even potential targetable mutations that are actionable targets in other malignancies have not shown the appropriate responses in clinical trials for bone sarcomas. Investigations of new systemic therapy, including molecular targeted therapies for bone sarcomas, have thus not progressed like those for other solid tumors. Another problem is that high rates of pediatric/adolescent and young adult patients have bone sarcomas such as osteosarcoma, and patient recruitment for clinical trials (especially randomized trials) is challenging. For pediatric patients, evaluations of tolerability and appropriate dose modifications of new drugs are needed, as their findings could provide the threshold for investigating new drugs for bone sarcomas. To solve these problems, improvements in registry systems, real world data, and pediatric extrapolation have been attempted. We review the issues regarding targeted drug investigations for bone sarcomas, focusing on the current clinical evidence and efforts to resolve these issues.
Collapse
|
38
|
Tarantino P, Carmagnani Pestana R, Corti C, Modi S, Bardia A, Tolaney SM, Cortes J, Soria JC, Curigliano G. Antibody-drug conjugates: Smart chemotherapy delivery across tumor histologies. CA Cancer J Clin 2022; 72:165-182. [PMID: 34767258 DOI: 10.3322/caac.21705] [Citation(s) in RCA: 175] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/26/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
As distinct cancer biomarkers have been discovered in recent years, a need to reclassify tumors by more than their histology has been proposed, and therapies are now tailored to treat cancers based on specific molecular aberrations and immunologic markers. In fact, multiple histology-agnostic therapies are currently adopted in clinical practice for treating patients regardless of their tumor site of origin. In parallel with this new model for drug development, in the past few years, several novel antibody-drug conjugates (ADCs) have been approved to treat solid tumors, benefiting from engineering improvements in the conjugation process and the introduction of novel linkers and payloads. With the recognition that numerous surface targets are expressed across various cancer histologies, alongside the remarkable activity of modern ADCs, this drug class has been increasingly evaluated as suitable for a histology-agnostic expansion of indication. For illustration, the anti-HER2 ADC trastuzumab deruxtecan has demonstrated compelling activity in HER2-overexpressing breast, gastric, colorectal, and lung cancer. Examples of additional novel and potentially histology-agnostic ADC targets include trophoblast cell-surface antigen 2 (Trop-2) and nectin-4, among others. In the current review article, the authors summarize the current approvals of ADCs by the US Food and Drug Administration focusing on solid tumors and discuss the challenges and opportunities posed by the multihistological expansion of ADCs.
Collapse
Affiliation(s)
- Paolo Tarantino
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Roberto Carmagnani Pestana
- Dayan-Daycoval Family Center for Oncology and Hematology, Albert Einstein Israelite Hospital, Sao Paulo, Brazil
| | - Chiara Corti
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Shanu Modi
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | - Aditya Bardia
- Harvard Medical School, Boston, Massachusetts
- Breast Cancer Treatment Program, Massachusetts General Hospital, Boston, Massachusetts
| | - Sara M Tolaney
- Harvard Medical School, Boston, Massachusetts
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Javier Cortes
- International Breast Cancer Center, Quironsalud Group, Barcelona, Spain
- Medica Scientia Innovation Research, Barcelona, Spain
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, European University of Madrid, Madrid, Spain
| | - Jean-Charles Soria
- Paris Saclay University, St Aubin, France
- Drug Development Department, Gustave Roussy, Villejuif, France
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
39
|
Zabor EC, Kane MJ, Roychoudhury S, Nie L, Hobbs BP. Bayesian basket trial design with false-discovery rate control. Clin Trials 2022; 19:297-306. [PMID: 35128970 DOI: 10.1177/17407745211073624] [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: 11/17/2022]
Abstract
BACKGROUND Recent advances in developing "tumor agnostic" oncology therapies have identified molecular targets that define patient subpopulations in a manner that supersedes conventional criteria for cancer classification. These successes have produced effective targeted therapies that are administered to patients regardless of their tumor histology. Trials have evolved as well with master protocol designs. By blending translational and clinical science, basket trials in particular are well-suited to investigate and develop targeted therapies among multiple cancer histologies. However, basket trials intrinsically involve more complex design decisions, including issues of multiple testing across baskets, and guidance for investigators is needed. METHODS The sensitivity of the multisource exchangeability model to prior specification under differing degrees of response heterogeneity is explored through simulation. Then, a multisource exchangeability model design that incorporates control of the false-discovery rate is presented and a simulation study compares the operating characteristics to a design where the family-wise error rate is controlled and to the frequentist approach of treating the baskets as independent. Simulations are based on the original design of a real-world clinical trial, the SUMMIT trial, which investigated Neratinib treatment for a variety of solid tumors. The methods studied here are specific to single-arm phase II trials with binary outcomes. RESULTS Values of prior probability of exchangeability in the multisource exchangeability model between 0.1 and 0.3 provide the best trade-offs between gain in precision and bias, especially when per-basket sample size is below 30. Application of these calibration results to a re-analysis of the SUMMIT trial showed that the breast basket exceeded the null response rate with posterior probability of 0.999 while having low posterior probability of exchangeability with all other baskets. Simulations based on the design of the SUMMIT trial revealed that there is meaningful improvement in power even in baskets with small sample size when the false-discovery rate is controlled as opposed to the family-wise error rate. For example, when only the breast basket was active, with a sample size of 25, the power was 0.76 when the false-discovery rate was controlled at 0.05 but only 0.56 when the family-wise error rate was controlled at 0.05, indicating that impractical sample sizes for the phase II setting would be needed to achieve acceptable power while controlling the family-wise error rate in this setting of a trial with 10 baskets. CONCLUSION Selection of the prior exchangeability probability based on calibration and incorporation of false-discovery rate control result in multisource exchangeability model designs with high power to detect promising treatments in the context of phase II basket trials.
Collapse
Affiliation(s)
| | | | | | - Lei Nie
- U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Brian P Hobbs
- Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
40
|
Aida Y, Fukushima H, Bando H, Ishikawa E, Shikama A, Sekine I. Characteristics of Cancers in Adolescents and Young Adults Compared with Those in Adults in Their 60s: A Single-Center Experience. Oncology 2022; 100:140-147. [PMID: 35100598 DOI: 10.1159/000520291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/17/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Cancer is one of the main causes of death among adolescents and young adults (AYAs) aged 15-39 years. The improvement in overall 5-year survival in AYA cancer patients was far below than that of adult cancer patients. The purpose of this study was to clarify the features of cancer in AYAs by comparing them with those of controls. METHODS Patients in the cancer registry of the University of Tsukuba Hospital between 2007 and 2017 (median age, 65 years) were included in this study. We used patients between the ages of 64 and 66 years as controls. We then obtained the age at diagnosis, sex, primary site, and pathological type. RESULTS Among 27,281 cancer patients in the registry between 2007 and 2017, 1,947 (7.1%) patients were categorized into the AYA group, and 2,354 into the control group. Among men in the AYA group, central nervous system (CNS) tumors accounted for 22.7% of all cancers, followed by germ cell tumors, 22.5%, and hematopoietic malignancies, 12.5%. Among women in the AYA group, cervical cancer accounted for 35.9% of all cancers, followed by breast cancer, 14.6%, and CNS tumors, 11.6%. The proportion of specific cancer types relative to all cancers in the CNS, thyroid, adrenal glands, germ cells, cervix uteri, hematopoietic tissues, and sarcomas was higher in the AYA group than that in the control group. CONCLUSION The present results for AYAs were in sharp contrast to those for adult cancers and may be related to different modes of pathogenesis in AYAs. The identification of high-risk groups of these tumors in the AYA generation is crucial for prevention and early detection and will be a major topic for future research. While most of adult cancers are treated independently by each medical department, AYA cancers need to be treated in collaboration with experts from several departments. It is desirable to address the issues involved in applying treatments established for adult cancers to AYA cancers on a cancer-by-cancer basis.
Collapse
Affiliation(s)
- Yuka Aida
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroko Fukushima
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroko Bando
- Department of Breast and Endocrine Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Eiichi Ishikawa
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ayumi Shikama
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ikuo Sekine
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
41
|
Jiang Y, Chan CKW, Chan RCK, Wang X, Wong N, To KF, Ng SSM, Lau JYW, Poon CCY. Identification of Tissue Types and Gene Mutations From Histopathology Images for Advancing Colorectal Cancer Biology. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2022; 3:115-123. [PMID: 35937101 PMCID: PMC9355144 DOI: 10.1109/ojemb.2022.3192103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/12/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Objective: Colorectal cancer (CRC) patients respond differently to treatments and are sub-classified by different approaches. We evaluated a deep learning model, which adopted endoscopic knowledge learnt from AI-doscopist, to characterise CRC patients by histopathological features. Results: Data of 461 patients were collected from TCGA-COAD database. The proposed framework was able to 1) differentiate tumour from normal tissues with an Area Under Receiver Operating Characteristic curve (AUROC) of 0.97; 2) identify certain gene mutations (MYH9, TP53) with an AUROC > 0.75; 3) classify CMS2 and CMS4 better than the other subtypes; and 4) demonstrate the generalizability of predicting KRAS mutants in an external cohort. Conclusions: Artificial intelligent can be used for on-site patient classification. Although KRAS mutants were commonly associated with therapeutic resistance and poor prognosis, subjects with predicted KRAS mutants in this study have a higher survival rate in 30 months after diagnoses.
Collapse
Affiliation(s)
- Yuqi Jiang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR
| | - Cecilia K. W. Chan
- Division of Vascular and General Surgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR
| | - Ronald C. K. Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR
| | - Nathalie Wong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Simon S. M. Ng
- Division of Colorectal Surgery, Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR
| | - James Y. W. Lau
- Division of Vascular and General Surgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR
| | | |
Collapse
|
42
|
Pestana RC, Beal JR, Parkes A, Hamerschlak N, Subbiah V. Impact of tissue-agnostic approvals for patients with sarcoma. Trends Cancer 2021; 8:135-144. [PMID: 34887213 DOI: 10.1016/j.trecan.2021.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022]
Abstract
Tissue-agnostic drug development is a major step forward in offering treatment options for rare tumors. Sarcomas are heterogeneous rare malignancies with more than 100 subtypes. Recent failure of Phase III trials, nonbiomarker-driven clinical trials, and rarity hamper developmental therapeutics in sarcomas. Since a 'one-size-fits-all' approach continues to be the standard of care, tissue-agnostic approvals assume significance in sarcomas. In this review, we focus on the clinical evidence of recent drug approvals for neurotrophic tyrosine receptor kinase (NTRK) fusion, microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) phenotype, and tumor mutation burden-high (TMB-H) status in the context of sarcomas, and the future landscape of tissue-agnostic targets, such as rearranged during transfection (RET), fibroblast growth factor receptor (FGFR), and neuregulin-1 (NRG1).
Collapse
Affiliation(s)
- Roberto Carmagnani Pestana
- Centro de Oncologia e Hematologia Família Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil.
| | - Juliana Rodrigues Beal
- Centro de Oncologia e Hematologia Família Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Amanda Parkes
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Nelson Hamerschlak
- Centro de Oncologia e Hematologia Família Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; MD Anderson Cancer Network, The University of Texas MD Anderson Cancer Center.
| |
Collapse
|
43
|
Zhuo LS, Wang MS, Wu FX, Xu HC, Gong Y, Yu ZC, Tian YG, Pang C, Hao GF, Huang W, Yang GF. Discovery of Next-Generation Tropomyosin Receptor Kinase Inhibitors for Combating Multiple Resistance Associated with Protein Mutation. J Med Chem 2021; 64:15503-15514. [PMID: 34668694 DOI: 10.1021/acs.jmedchem.1c01539] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tropomyosin receptor kinase (TRK) inhibition is an effective therapeutic approach for treatment of a variety of cancers. Despite the use of first-generation TRK inhibitor (TRKI) larotrectinib (1) resulting in significant therapeutic response in patients, acquired resistance develops invariably. The emergence of secondary mutations occurring at the solvent-front, xDFG, and gatekeeper regions of TRK represents a common mechanism for acquired resistance. However, xDFG mutations remain insensitive to second-generation macrocyclic TRKIs selitrectinib (3) and repotrectinib (4) designed to overcome the resistance mediated by solvent-front and gatekeeper mutations. Here, we report the structure-based drug design and discovery of a next-generation TRKI. The structure-activity relationship studies culminated in the identification of a promising drug candidate 8 that showed excellent in vitro potency on a panel of TRK mutants, especially TRKAG667C in the xDFG motif, and improved in vivo efficacy than 1 and 3 in TRK wild-type and mutant fusion-driven tumor xenograft models, respectively.
Collapse
Affiliation(s)
- Lin-Sheng Zhuo
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Ming-Shu Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Feng-Xu Wu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Hubei 442000, P. R. China
| | - Hong-Chuang Xu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Yi Gong
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Zhi-Cheng Yu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Yan-Guang Tian
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Chao Pang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Ge-Fei Hao
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Wei Huang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China
| |
Collapse
|
44
|
Cheng P, Shen P, Shan Y, Yang Y, Deng R, Chen W, Lu Y, Wei Z. Gut Microbiota-Mediated Modulation of Cancer Progression and Therapy Efficacy. Front Cell Dev Biol 2021; 9:626045. [PMID: 34568308 PMCID: PMC8455814 DOI: 10.3389/fcell.2021.626045] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 08/18/2021] [Indexed: 12/18/2022] Open
Abstract
The role of gut microbiota in the development of various tumors has been a rising topic of public interest, and in recent years, many studies have reported a close relationship between microbial groups and tumor development. Gut microbiota play a role in host metabolism, and the positive and negative alterations of these microbiota have an effect on tumor treatment. The microbiota directly promote, eliminate, and coordinate the efficacy of chemotherapy drugs and the toxicity of adjuvant drugs, and enhance the ability of patients to respond to tumors in adjuvant immunotherapy. In this review, we outline the significance of gut microbiota in tumor development, reveal its impacts on chemotherapy and immunotherapy, and discover various potential mechanisms whereby they influence tumor treatment. This review demonstrates the importance of intestinal microbiota-related research for clinical tumor treatment and provides additional strategy for clinical assistance in cancer treatment.
Collapse
Affiliation(s)
- Peng Cheng
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peiliang Shen
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yu Yang
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
45
|
Weis LN, Tolaney SM, Barrios CH, Barroso-Sousa R. Tissue-agnostic drug approvals: how does this apply to patients with breast cancer? NPJ Breast Cancer 2021; 7:120. [PMID: 34518552 PMCID: PMC8437983 DOI: 10.1038/s41523-021-00328-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/20/2021] [Indexed: 02/08/2023] Open
Abstract
Precision medicine has provided new perspectives in oncology, yielding research on the use of targeted therapies across different tumor types, regardless of their site of origin, a concept known as tissue-agnostic indication. Since 2017, the Food and Drug Administration (FDA) has approved the use of three different agents for tumor-agnostic treatment: pembrolizumab (for patients with microsatellite instability or high tumor mutational burden) and larotrectinib and entrectinib (both for use in patients harboring tumors with NTRK fusions). Importantly, the genomic alterations targeted by these agents are uncommon or rare in breast cancer, and little information exists regarding their efficacy in advanced breast cancer. In this review, we discuss the prevalence of these targets in breast cancer, their detection methods, the clinical characteristics of patients whose tumors have these alterations, and available data regarding the efficacy of these agents in breast cancer.
Collapse
Affiliation(s)
- Luiza N Weis
- Instituto de Ensino e Pesquisa Hospital Sírio-Libanês, São Paulo-SP, Brazil
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Romualdo Barroso-Sousa
- Instituto de Ensino e Pesquisa Hospital Sírio-Libanês, São Paulo-SP, Brazil.
- Oncology Center, Hospital Sírio-Libanês Brasília, Brasília-DF, Brazil.
| |
Collapse
|
46
|
Wu J, Li C, Gensheimer M, Padda S, Kato F, Shirato H, Wei Y, Schönlieb CB, Price SJ, Jaffray D, Heymach J, Neal JW, Loo BW, Wakelee H, Diehn M, Li R. Radiological tumor classification across imaging modality and histology. NAT MACH INTELL 2021; 3:787-798. [PMID: 34841195 PMCID: PMC8612063 DOI: 10.1038/s42256-021-00377-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Radiomics refers to the high-throughput extraction of quantitative features from radiological scans and is widely used to search for imaging biomarkers for prediction of clinical outcomes. Current radiomic signatures suffer from limited reproducibility and generalizability, because most features are dependent on imaging modality and tumor histology, making them sensitive to variations in scan protocol. Here, we propose novel radiological features that are specially designed to ensure compatibility across diverse tissues and imaging contrast. These features provide systematic characterization of tumor morphology and spatial heterogeneity. In an international multi-institution study of 1,682 patients, we discover and validate four unifying imaging subtypes across three malignancies and two major imaging modalities. These tumor subtypes demonstrate distinct molecular characteristics and prognoses after conventional therapies. In advanced lung cancer treated with immunotherapy, one subtype is associated with improved survival and increased tumor-infiltrating lymphocytes compared with the others. Deep learning enables automatic tumor segmentation and reproducible subtype identification, which can facilitate practical implementation. The unifying radiological tumor classification may inform prognosis and treatment response for precision medicine.
Collapse
Affiliation(s)
- Jia Wu
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic and Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Chao Li
- The Centre for Mathematical Imaging in Healthcare, Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, UK
- Cambridge Brain Tumor Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Michael Gensheimer
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sukhmani Padda
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fumi Kato
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Hiroki Shirato
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yiran Wei
- Cambridge Brain Tumor Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Stephen John Price
- Cambridge Brain Tumor Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - David Jaffray
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
- Office of the Chief Technology and Digital Officer, MD Anderson Cancer Center, Houston, TX, USA
| | - John Heymach
- Department of Thoracic and Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Joel W Neal
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Heather Wakelee
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
47
|
Zhou J, Jiang X, Amy Xia H, Wei P, Hobbs BP. A survival mediation model with Bayesian model averaging. Stat Methods Med Res 2021; 30:2413-2427. [PMID: 34448657 DOI: 10.1177/09622802211037069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Determining the extent to which a patient is benefiting from cancer therapy is challenging. Criteria for quantifying the extent of "tumor response" observed within a few cycles of treatment have been established for various types of solids as well as hematologic malignancies. These measures comprise the primary endpoints of phase II trials. Regulatory approvals of new cancer therapies, however, are usually contingent upon the demonstration of superior overall survival with randomized evidence acquired with a phase III trial comparing the novel therapy to an appropriate standard of care treatment. With nearly two-thirds of phase III oncology trials failing to achieve statistically significant results, researchers continue to refine and propose new surrogate endpoints. This article presents a Bayesian framework for studying relationships among treatment, patient subgroups, tumor response, and survival. Combining classical components of a mediation analysis with Bayesian model averaging, the methodology is robust to model misspecification among various possible relationships among the observable entities. A posterior inference is demonstrated via an application to a randomized controlled phase III trial in metastatic colorectal cancer. Moreover, the article details posterior predictive distributions of survival and statistical metrics for quantifying the extent of direct and indirect, or tumor response mediated treatment effects.
Collapse
Affiliation(s)
- Jie Zhou
- Quantitative Health Sciences, 2569Cleveland Clinic, USA
| | - Xun Jiang
- 13498Center for Design and Analysis, Amgen, USA
| | | | - Peng Wei
- 4002The University of Texas MD Anderson Cancer Center, USA
| | - Brian P Hobbs
- Dell Medical School, 12330The University of Texas at Austin, USA
| |
Collapse
|
48
|
Kim JH, Hong JH, Choi YL, Lee JA, Seo MK, Lee MS, An SB, Sung MJ, Cho NY, Kim SS, Shin YK, Kim S, Kang GH. NTRK oncogenic fusions are exclusively associated with the serrated neoplasia pathway in the colorectum and begin to occur in sessile serrated lesions. J Pathol 2021; 255:399-411. [PMID: 34402529 DOI: 10.1002/path.5779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/28/2021] [Accepted: 08/13/2021] [Indexed: 01/16/2023]
Abstract
Neurotrophic tropomyosin receptor kinase (NTRK) gene fusions are emerging tissue-agnostic drug targets in malignancies including colorectal carcinomas (CRCs), but their detailed landscape in the context of various colorectal carcinogenesis pathways remains to be investigated. In this study, pan-tropomyosin receptor kinase (TRK) protein expression was assessed by immunohistochemistry (IHC) in retrospectively collected colorectal epithelial tumor tissues, including 441 CRCs [133 microsatellite instability-high (MSI-high) and 308 microsatellite stable (MSS)] and 595 premalignant colorectal lesions (330 serrated lesions and 265 conventional adenomas). TRK-positive cases were then subjected to next-generation sequencing and/or fluorescence in situ hybridization to confirm NTRK rearrangements. TRK IHC positivity was not observed in any of the MSS CRCs, conventional adenomas, traditional serrated adenomas, or hyperplastic polyps, whereas TRK positivity was observed in 11 of 58 (19%) MLH1-methylated MSI-high CRCs, 4 of 23 (17%) sessile serrated lesions with dysplasia (SSLDs), and 5 of 132 (4%) sessile serrated lesions (SSLs). The 11 TRK-positive MSI-high CRCs commonly harbored CpG island methylator phenotype-high (CIMP-high), MLH1 methylation, BRAF/KRAS wild-type, and NTRK1 or NTRK3 fusion (TPM3-NTRK1, TPR-NTRK1, LMNA-NTRK1, SFPQ-NTRK1, ETV6-NTRK3, or EML4-NTRK3). Both NTRK1 or NTRK3 rearrangement and BRAF/KRAS wild-type were detected in all nine TRK-positive SSL(D)s, seven of which demonstrated MSS and/or CIMP-low. TRK expression was selectively observed in distorted serrated crypts within SSLs and was occasionally localized at the base of serrated crypts. NTRK fusions were detected only in SSLs of patients aged ≥50 years, whereas BRAF mutation was found in younger age-onset SSLs. In conclusion, NTRK-rearranged colorectal tumors develop exclusively through the serrated neoplasia pathway and can be initiated from non-dysplastic SSLs without BRAF/KRAS mutations prior to full occurrence of MSI-high/CIMP-high. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jung Ho Kim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeong Hoon Hong
- Central Laboratory, LOGONE Bio-Convergence Research Foundation, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea.,Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Pathology and Translational Genomics, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ji Ae Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Mi-Kyoung Seo
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi-Sook Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea.,Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Bin An
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea.,Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Min Jung Sung
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea.,Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Nam-Yun Cho
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Su Kim
- Central Laboratory, LOGONE Bio-Convergence Research Foundation, Seoul, Republic of Korea
| | - Young Kee Shin
- Central Laboratory, LOGONE Bio-Convergence Research Foundation, Seoul, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
49
|
Orlov-Slavu MC, Popa AM, Tulin A, Pantea Stoian A, Poiana C, Paleru C, Calu V, Nitipir C. The Utility of Next-Generation Sequencing in the Treatment Decision-Making for Metastatic Non-Small-Cell Lung Cancer. Cureus 2021; 13:e16919. [PMID: 34513491 PMCID: PMC8412888 DOI: 10.7759/cureus.16919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 02/01/2023] Open
Abstract
Next-generation sequencing (NGS) is a fast and relatively inexpensive method to sequence a large number of genes with crucial importance in cancer medicine. Nowadays, NGS is frequently used in diagnostic and therapeutic decisions in oncology; however, recently, it was demonstrated that only a few cancer sites actually benefit from this assessment. Moreover, the association of a mutant gene with a targeted drug is not always as predicted during in-vitro trials and is often not associated with tumor response. To predict the efficacy of such an association several classification systems have been developed. The present review aims to analyze the most important tumor agnostic treatment trials and assess how they shape selecting cancer patients for NGS. Moreover, it aims to determine how mutation-drug associations can be classified by their targetability and level of evidence of efficacy in non-small-cell lung cancer.
Collapse
Affiliation(s)
| | - Ana Maria Popa
- Oncology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Adrian Tulin
- General Surgery, Agrippa Ionescu Emergency Hospital, Bucharest, ROU
- Human Anatomy Department, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Anca Pantea Stoian
- Diabetes, Nutrition and Metabolic Diseases Department, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Catalina Poiana
- Endocrinology, National Institute of Endocrinology C.I.Parhon, Bucharest, ROU
| | - Cristian Paleru
- Thoracic Surgery, Marius Nasta National Institute of Pneumology, Bucharest, ROU
| | - Valentin Calu
- General Surgery, Elias Emergency Hospital, Bucharest, ROU
| | - Cornelia Nitipir
- Oncology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| |
Collapse
|
50
|
Minami H, Kiyota N, Kimbara S, Ando Y, Shimokata T, Ohtsu A, Fuse N, Kuboki Y, Shimizu T, Yamamoto N, Nishio K, Kawakami Y, Nihira SI, Sase K, Nonaka T, Takahashi H, Komori Y, Kiyohara K. Guidelines for clinical evaluation of anti-cancer drugs. Cancer Sci 2021; 112:2563-2577. [PMID: 33990993 PMCID: PMC8253284 DOI: 10.1111/cas.14967] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Clinical studies intended for regulatory approval must demonstrate the clinical benefits of the drug in a target population. Clinical development of a drug proceeds by stepwise clinical studies; after safety and pharmacokinetics are evaluated and the recommended dosage and administration are determined, efficacy and safety are evaluated in an exploratory manner, and finally clinical benefits are compared with conventional standard therapies. Guidelines for the clinical evaluation of anti‐cancer drugs in Japan were established in 1991 and amended in 2006 after molecular‐targeted drugs were introduced. Recent progress in the development of drugs acting on the immune system and cancer genomic medicine targeting rare but important molecular subtypes have altered the strategy for development of anti‐cancer drugs. It is often difficult to conduct a confirmatory randomized controlled study using overall survival as the primary endpoint in rare molecular subtypes, and the primary evaluation of the efficacy of some drugs and subsequent approval is based on the tumor response. As conducting clinical studies for rare subtypes solely within Japan is difficult, drug development needs to be conducted within a global study. However, this requires robust monitoring to detect possible ethnic differences in pharmacokinetics and drug efficacy. Development using the conditional approval system for drugs enforced in 2020 may be considered, when clinical utility is evaluated based on surrogate endpoints. Because of these changes, we have revised the guidelines for the clinical evaluation of anti‐cancer drugs in Japan. To promote global development of anti‐cancer drugs involving Japan, the guidelines have been translated into English. Recent progress in development of drugs acting on the immune system and cancer genomic medicine targeting rare but important molecular subtypes have altered the strategy for development of anti‐cancer drugs. As conducting clinical studies for rare subtypes solely within Japan is difficult, drug development needs to be conducted within a global study with monitoring possible ethnic differences. Because of these changes, we have revised the guidelines for the clinical evaluation of anti‐cancer drugs in Japan.
Collapse
Affiliation(s)
- Hironobu Minami
- Division of Medical Oncology and Hematology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.,Cancer Center, Kobe University Hospital, Kobe, Japan
| | - Naomi Kiyota
- Division of Medical Oncology and Hematology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.,Cancer Center, Kobe University Hospital, Kobe, Japan
| | - Shiro Kimbara
- Division of Medical Oncology and Hematology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Tomoya Shimokata
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Atsushi Ohtsu
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Nozomu Fuse
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yasutoshi Kuboki
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toshio Shimizu
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yutaka Kawakami
- Department of Immunology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Shin-Ichi Nihira
- Tokyo Biochemical Research Foundation-Comprehensive Academy for Advanced Oncology, Tokyo, Japan
| | - Kazuhiro Sase
- Clinical Pharmacology & Regulatory Science, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takahiro Nonaka
- Division of Epidemiology, Office of Medical Informatics and Epidemiology, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Hideaki Takahashi
- Office of New Drug V, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Yukiko Komori
- Office of New Drug IV, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Koshin Kiyohara
- Office of New Drug V, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| |
Collapse
|