Your browser doesn't support javascript.
loading
Single-cell analysis of treatment-resistant prostate cancer: Implications of cell state changes for cell surface antigen-targeted therapies.
Zaidi, Samir; Park, Jooyoung; Chan, Joseph M; Roudier, Martine P; Zhao, Jimmy L; Gopalan, Anuradha; Wadosky, Kristine M; Patel, Radhika A; Sayar, Erolcan; Karthaus, Wouter R; Kates, D Henry; Chaudhary, Ojasvi; Xu, Tianhao; Masilionis, Ignas; Mazutis, Linas; Chaligné, Ronan; Obradovic, Aleksandar; Linkov, Irina; Barlas, Afsar; Jungbluth, Achim A; Rekhtman, Natasha; Silber, Joachim; Manova-Todorova, Katia; Watson, Philip A; True, Lawrence D; Morrissey, Colm; Scher, Howard I; Rathkopf, Dana E; Morris, Michael J; Goodrich, David W; Choi, Jungmin; Nelson, Peter S; Haffner, Michael C; Sawyers, Charles L.
Afiliação
  • Zaidi S; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Park J; Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Chan JM; Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea.
  • Roudier MP; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Zhao JL; Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Gopalan A; Department of Urology, University of Washington, Seattle, WA 98195.
  • Wadosky KM; Astrazeneca Oncology R&D, New York, NY 10016.
  • Patel RA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Sayar E; Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263.
  • Karthaus WR; Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98195.
  • Kates DH; Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA 98195.
  • Chaudhary O; Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98195.
  • Xu T; Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA 98195.
  • Masilionis I; Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland.
  • Mazutis L; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Chaligné R; Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Obradovic A; Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Linkov I; Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Barlas A; Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Jungbluth AA; Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Rekhtman N; Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032.
  • Silber J; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Manova-Todorova K; Molecular Cytology Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York NY 10065.
  • Watson PA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • True LD; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Morrissey C; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Scher HI; Molecular Cytology Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York NY 10065.
  • Rathkopf DE; Research Outreach and Compliance, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Morris MJ; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195.
  • Goodrich DW; Department of Urology, University of Washington, Seattle, WA 98195.
  • Choi J; Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Nelson PS; Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Haffner MC; Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • Sawyers CL; Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263.
Proc Natl Acad Sci U S A ; 121(28): e2322203121, 2024 Jul 09.
Article em En | MEDLINE | ID: mdl-38968122
ABSTRACT
Targeting cell surface molecules using radioligand and antibody-based therapies has yielded considerable success across cancers. However, it remains unclear how the expression of putative lineage markers, particularly cell surface molecules, varies in the process of lineage plasticity, wherein tumor cells alter their identity and acquire new oncogenic properties. A notable example of lineage plasticity is the transformation of prostate adenocarcinoma (PRAD) to neuroendocrine prostate cancer (NEPC)-a growing resistance mechanism that results in the loss of responsiveness to androgen blockade and portends dismal patient survival. To understand how lineage markers vary across the evolution of lineage plasticity in prostate cancer, we applied single-cell analyses to 21 human prostate tumor biopsies and two genetically engineered mouse models, together with tissue microarray analysis on 131 tumor samples. Not only did we observe a higher degree of phenotypic heterogeneity in castrate-resistant PRAD and NEPC than previously anticipated but also found that the expression of molecules targeted therapeutically, namely PSMA, STEAP1, STEAP2, TROP2, CEACAM5, and DLL3, varied within a subset of gene-regulatory networks (GRNs). We also noted that NEPC and small cell lung cancer subtypes shared a set of GRNs, indicative of conserved biologic pathways that may be exploited therapeutically across tumor types. While this extreme level of transcriptional heterogeneity, particularly in cell surface marker expression, may mitigate the durability of clinical responses to current and future antigen-directed therapies, its delineation may yield signatures for patient selection in clinical trials, potentially across distinct cancer types.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Análise de Célula Única Limite: Animals / Humans / Male Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Análise de Célula Única Limite: Animals / Humans / Male Idioma: En Ano de publicação: 2024 Tipo de documento: Article