Your browser doesn't support javascript.
loading
Minimal Residual Disease in Myeloma: Application for Clinical Care and New Drug Registration.
Anderson, Kenneth C; Auclair, Daniel; Adam, Stacey J; Agarwal, Amit; Anderson, Melissa; Avet-Loiseau, Hervé; Bustoros, Mark; Chapman, Jessica; Connors, Dana E; Dash, Ajeeta; Di Bacco, Alessandra; Du, Ling; Facon, Thierry; Flores-Montero, Juan; Gay, Francesca; Ghobrial, Irene M; Gormley, Nicole J; Gupta, Ira; Higley, Howard; Hillengass, Jens; Kanapuru, Bindu; Kazandjian, Dickran; Kelloff, Gary J; Kirsch, Ilan R; Kremer, Brandon; Landgren, Ola; Lightbody, Elizabeth; Lomas, Oliver C; Lonial, Sagar; Mateos, María-Victoria; Montes de Oca, Rocio; Mukundan, Lata; Munshi, Nikhil C; O'Donnell, Elizabeth K; Orfao, Alberto; Paiva, Bruno; Patel, Reshma; Pugh, Trevor J; Ramasamy, Karthik; Ray, Jill; Roshal, Mikhail; Ross, Jeremy A; Sigman, Caroline C; Thoren, Katie L; Trudel, Suzanne; Ulaner, Gary; Valente, Nancy; Weiss, Brendan M; Zamagni, Elena; Kumar, Shaji K.
Afiliação
  • Anderson KC; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Auclair D; Multiple Myeloma Research Foundation, Norwalk, Connecticut.
  • Adam SJ; Foundation for the National Institutes of Health, North Bethesda, Maryland.
  • Agarwal A; US Medical Oncology, Bristol-Myers Squibb, Summit, New Jersey.
  • Anderson M; Takeda Pharmaceuticals, Cambridge, Massachusetts.
  • Avet-Loiseau H; Laboratoire d'Hématologie, Pôle Biologie, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France.
  • Bustoros M; Division of Hematology and Medical Oncology, Cornell University/New York Presbyterian Hospital, New York, New York.
  • Chapman J; Memorial Sloan Kettering Cancer Center, New York, New York.
  • Connors DE; Foundation for the National Institutes of Health, North Bethesda, Maryland.
  • Dash A; Takeda Pharmaceuticals, Cambridge, Massachusetts.
  • Di Bacco A; Takeda Pharmaceuticals, Cambridge, Massachusetts.
  • Du L; GlaxoSmithKline, Collegeville, Pennsylvania.
  • Facon T; Department of Hematology, Lille University Hospital, Lille, France.
  • Flores-Montero J; Cancer Research Center (IBMCC-CSIC/USAL-IBSAL); Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Salamanca, Spain.
  • Gay F; Myeloma Unit, Division of Hematology, Azienda Ospedaliero Università Città della Salute e della Scienza, Torino, Italy.
  • Ghobrial IM; Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Gormley NJ; Division of Hematologic Malignancies 2, Office of Oncologic Disease, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland.
  • Gupta I; GlaxoSmithKline, Collegeville, Pennsylvania.
  • Higley H; CCS Associates, Inc., San Jose, California.
  • Hillengass J; Division of Hematology and Oncology, Roswell Park Cancer Institute, Buffalo, New York.
  • Kanapuru B; Division of Hematologic Malignancies 2, Office of Oncologic Disease, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland.
  • Kazandjian D; Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.
  • Kelloff GJ; Division of Cancer Treatment and Diagnosis, NCI, NIH, Rockville, Maryland.
  • Kirsch IR; Translational Medicine, Adaptive Biotechnologies, Seattle, Washington.
  • Kremer B; GlaxoSmithKline, Collegeville, Pennsylvania.
  • Landgren O; Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.
  • Lightbody E; Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Lomas OC; Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Lonial S; Department of Hematology and Medical Oncology at Emory University School of Medicine, Atlanta, Georgia.
  • Mateos MV; Haematology Department, University of Salamanca, Salamanca, Spain.
  • Montes de Oca R; GlaxoSmithKline, Collegeville, Pennsylvania.
  • Mukundan L; CCS Associates, Inc., San Jose, California.
  • Munshi NC; Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • O'Donnell EK; Multiple Myeloma Disease Center, Massachusetts General Hospital, Boston, Massachusetts.
  • Orfao A; Cancer Research Center (IBMCC-CSIC/USAL-IBSAL); Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Salamanca, Spain.
  • Paiva B; Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain.
  • Patel R; Janssen Research & Development, Spring House, Pennsylvania.
  • Pugh TJ; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
  • Ramasamy K; Cancer and Haematology Centre, Oxford University Hospitals, Oxford, United Kingdom.
  • Ray J; BioOncology, Genentech Inc., South San Francisco, California.
  • Roshal M; Memorial Sloan Kettering Cancer Center, New York, New York.
  • Ross JA; Precision Medicine, Oncology, AbbVie, Inc., North Chicago, Illinois.
  • Sigman CC; CCS Associates, Inc., San Jose, California.
  • Thoren KL; Memorial Sloan Kettering Cancer Center, New York, New York.
  • Trudel S; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
  • Ulaner G; Hoag Cancer Center, Irvine, California.
  • Valente N; BioOncology, Genentech Inc., South San Francisco, California.
  • Weiss BM; Janssen Research & Development, Spring House, Pennsylvania.
  • Zamagni E; Seragnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy.
  • Kumar SK; Division of Hematology, Mayo Clinic, Rochester, Minnesota.
Clin Cancer Res ; 27(19): 5195-5212, 2021 10 01.
Article em En | MEDLINE | ID: mdl-34321279
ABSTRACT
The development of novel agents has transformed the treatment paradigm for multiple myeloma, with minimal residual disease (MRD) negativity now achievable across the entire disease spectrum. Bone marrow-based technologies to assess MRD, including approaches using next-generation flow and next-generation sequencing, have provided real-time clinical tools for the sensitive detection and monitoring of MRD in patients with multiple myeloma. Complementary liquid biopsy-based assays are now quickly progressing with some, such as mass spectrometry methods, being very close to clinical use, while others utilizing nucleic acid-based technologies are still developing and will prove important to further our understanding of the biology of MRD. On the regulatory front, multiple retrospective individual patient and clinical trial level meta-analyses have already shown and will continue to assess the potential of MRD as a surrogate for patient outcome. Given all this progress, it is not surprising that a number of clinicians are now considering using MRD to inform real-world clinical care of patients across the spectrum from smoldering myeloma to relapsed refractory multiple myeloma, with each disease setting presenting key challenges and questions that will need to be addressed through clinical trials. The pace of advances in targeted and immune therapies in multiple myeloma is unprecedented, and novel MRD-driven biomarker strategies are essential to accelerate innovative clinical trials leading to regulatory approval of novel treatments and continued improvement in patient outcomes.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Mieloma Múltiplo Tipo de estudo: Diagnostic_studies / Observational_studies Limite: Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Mieloma Múltiplo Tipo de estudo: Diagnostic_studies / Observational_studies Limite: Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article