1
|
Kale R, Samant C, Nandakumar K, Ranganath Pai KS, Bhonde M. Drugging the Undruggable and beyond: Emerging precision oncology approaches to target acquired resistance to KRAS G12C and KRAS G12D inhibitors. Biochem Biophys Res Commun 2025; 760:151688. [PMID: 40174369 DOI: 10.1016/j.bbrc.2025.151688] [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: 11/22/2024] [Revised: 02/21/2025] [Accepted: 03/22/2025] [Indexed: 04/04/2025]
Abstract
Development of mutant specific KRAS inhibitors validated KRAS as a 'druggable' target. However, excellent initial efficacy was eventually overshadowed by failure to exhibit sustained clinical response, primarily due to acquired resistance. Some targeted therapies like SOS1, SHP2, and MEK inhibitors, in combination with mutant KRAS G12C inhibitors (G12Ci), are currently under clinical investigation with evidences of improving efficacy. However, a deep understanding of the underlying molecular pathways behind the acquired resistance is still at a nascent stage. Recent preclinical studies have uncovered a role of novel proteins and pathways responsible for resistance and their inhibition demonstrated a robust anticancer efficacy in combination. Plethora of combination therapy approaches are now being proposed with emergence of AXL, ULK1, Tissue factor, farnesyltransferase, etc. as targets to counter G12Ci resistance. This review summarizes in a comprehensive manner, some of the novel combination modalities to overcome G12Ci resistance, based on current understanding and with great potential to hit clinical success. Along with G12C, KRAS G12D (G12D) was also considered a formidable foe, until the discovery of selective inhibitors. However, eventual clinical resistance can eclipse the early success and requires an in-depth understanding of resistance mechanisms. Evidences of G12Ci resistance can be exploited as probable combination strategies to tackle ensuing resistance to G12D inhibitors (G12Di), and can translate in superior clinical efficacy. Early preclinical studies of G12Di in combination with ERBB, SOS1, AKT and immune-checkpoints inhibitors indicate encouraging response. This review further describes some of the early affirmations on combination strategies with G12Di. We postulate to go beyond 'Drugging the Undruggable' with advanced combination approaches mitigating G12C and G12D inhibitor resistance.
Collapse
Affiliation(s)
- Ramesh Kale
- Research Scholar, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India; Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| | - Charudatt Samant
- Research Scholar, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India; Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India.
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Mandar Bhonde
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| |
Collapse
|
2
|
Yang P, Li Y. Progress of KRAS G12C inhibitors in the treatment of refractory colorectal cancer and strategies for drug resistance response. Invest New Drugs 2025; 43:357-364. [PMID: 39956882 DOI: 10.1007/s10637-025-01514-x] [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: 09/29/2024] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Colorectal cancer is the third most prevalent cancer in the world. Early screening and detection of tumours, active surgical radical treatment, postoperative adjuvant chemotherapy, targeted therapy, and immunotherapy are performed based on pathological staging and immunohistochemistry. Even with these measures, the 5-year survival rate of colorectal cancer is only 65%, and a considerable number of patients still experience tumour recurrence or even metastasis. The KRAS G12C mutation accounts for 3 to 4% of refractory colorectal cancer (advanced or metastatic colorectal cancer), and it was once believed that KRAS did not have a drug target until the emergence of KRAS G12C inhibitors provided targeted treatment for KRAS-mutated colorectal cancer. However, KRAS G12C inhibitors only produce moderate efficacy, and resistance occurs after a short remission. The mechanism of drug resistance in tumour cells is complex and diverse, and existing research has limited understanding of it. This review aims to elucidate the clinical trial progress of KRAS G12C inhibitors in refractory colorectal cancer, the research progress of drug resistance mechanisms, and the combined treatment strategies for drug resistance.
Collapse
Affiliation(s)
- Peiyuan Yang
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Xiantai Street, Changchun, 130000, Jilin, China
| | - Yongchao Li
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Xiantai Street, Changchun, 130000, Jilin, China.
| |
Collapse
|
3
|
Baltanás FC, Kramer-Drauberg M, García-Navas R, Patrucco E, Petrini E, Arnhof H, Olarte-San Juan A, Rodríguez-Ramos P, Borrajo J, Calzada N, Castellano E, Mair B, Kostyrko K, Hofmann MH, Ambrogio C, Santos E. SOS1 inhibitor BI-3406 shows in vivo antitumor activity akin to genetic ablation and synergizes with a KRAS G12D inhibitor in KRAS LUAD. Proc Natl Acad Sci U S A 2025; 122:e2422943122. [PMID: 40073053 PMCID: PMC11929440 DOI: 10.1073/pnas.2422943122] [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: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
We evaluated the in vivo therapeutic efficacy and tolerability of BI-3406-mediated pharmacological inhibition of SOS1 in comparison to genetic ablation of this universal Ras-GEF in various KRAS-dependent experimental tumor settings. Contrary to the rapid lethality caused by SOS1 genetic ablation in SOS2KO mice, SOS1 pharmacological inhibition by its specific inhibitor BI-3406 did not significantly affect animal weight/viability nor cause noteworthy systemic toxicity. Allograft assays using different KRASmut cell lines showed that treatment with BI-3406 impaired RAS activation and RAS downstream signaling and decreased tumor burden and disease progression as a result of both tumor-intrinsic and -extrinsic therapeutic effects of the drug. Consistent with prior genetic evidence and the KRASmut allografts assays in immunocompromised mice, our analyses using an in vivo model of KRASG12D-driven lung adenocarcinoma (LUAD) in immunocompetent mice showed that single, systemic BI-3406 treatment impaired tumor growth and downmodulated protumorigenic components of the tumor microenvironment comparably to SOS1 genetic ablation or to treatment with the specific KRASG12D inhibitor MRTX1133. Furthermore, markedly stronger, synergistic antitumor effects were observed upon concomitant treatment with BI-3406 and MRTX1133 in the same in vivo LUAD mouse model. Our data confirm SOS1 as an actionable therapy target in RAS-dependent cancers and suggest that BI-3406 treatment may yield clinical benefit both as monotherapy or as a potential combination partner for multiple RAS-targeting strategies.
Collapse
Affiliation(s)
- Fernando C. Baltanás
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla and Departamento de Fisiología Medica y Biofísica, Universidad de Sevilla, Sevilla41013, Spain
| | - Maximilian Kramer-Drauberg
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Rósula García-Navas
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Enrico Patrucco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Ettore Petrini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Heribert Arnhof
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Andrea Olarte-San Juan
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Pablo Rodríguez-Ramos
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Javier Borrajo
- Departamento de Ciencias Biomédicas y del Diagnóstico, Universidad de Salamanca, Salamanca37007, Spain
| | - Nuria Calzada
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Esther Castellano
- Laboratorio 5. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca37007, Spain
| | - Barbara Mair
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Kaja Kostyrko
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Marco H. Hofmann
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Eugenio Santos
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| |
Collapse
|
4
|
Ying H, Kimmelman AC, Bardeesy N, Kalluri R, Maitra A, DePinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 2025; 39:36-63. [PMID: 39510840 PMCID: PMC11789498 DOI: 10.1101/gad.351863.124] [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: 11/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a grim prognosis for patients. Recent multidisciplinary research efforts have provided critical insights into its genetics and tumor biology, creating the foundation for rational development of targeted and immune therapies. Here, we review the PDAC genomic landscape and the role of specific oncogenic events in tumor initiation and progression, as well as their contributions to shaping its tumor biology. We further summarize and synthesize breakthroughs in single-cell and metabolic profiling technologies that have illuminated the complex cellular composition and heterotypic interactions of the PDAC tumor microenvironment, with an emphasis on metabolic cross-talk across cancer and stromal cells that sustains anabolic growth and suppresses tumor immunity. These conceptual advances have generated novel immunotherapy regimens, particularly cancer vaccines, which are now in clinical testing. We also highlight the advent of KRAS targeted therapy, a milestone advance that has transformed treatment paradigms and offers a platform for combined immunotherapy and targeted strategies. This review provides a perspective summarizing current scientific and therapeutic challenges as well as practice-changing opportunities for the PDAC field at this major inflection point.
Collapse
Affiliation(s)
- Haoqiang Ying
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Alec C Kimmelman
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York 10016, USA
- Department of Radiation Oncology, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02114, USA
- The Cancer Program, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Raghu Kalluri
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Anirban Maitra
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Sheikh Ahmed Pancreatic Cancer Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA;
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
5
|
Finan JM, Guo Y, Goodyear SM, Brody JR. Challenges and Opportunities in Targeting the Complex Pancreatic Tumor Microenvironment. JCO ONCOLOGY ADVANCES 2024; 1:e2400050. [PMID: 39735733 PMCID: PMC11670921 DOI: 10.1200/oa-24-00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/16/2024] [Accepted: 11/04/2024] [Indexed: 12/31/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related deaths with a 5-year survival rate of 13%. Surgical resection remains the only curative option as systemic therapies offer limited benefit. Poor response to chemotherapy and immunotherapy is due, in part, to the dense stroma and heterogeneous tumor microenvironment (TME). Opportunities to target the PDAC stroma may increase the effectiveness of existing or novel therapies. Current strategies targeting the stromal compartment within the PDAC TME primarily focus on degrading extracellular matrix or inhibiting stromal cell activity, angiogenesis, or hypoxic responses. In addition, extensive work has attempted to use immune targeting strategies to improve clinical outcomes. Preclinically, these strategies show promise, especially with the ability to alter the tumor ecosystem; however, when translated to the clinic, most of these trials have failed to improve overall patient outcomes. In this review, we catalog the heterogenous elements of the TME and discuss the potential of combination therapies that target the heterogeneity observed in the TME between patients and how molecular stratification could improve responses to targeted and combination therapies.
Collapse
Affiliation(s)
- Jennifer M. Finan
- Department of Surgery, Oregon Health & Science University, Portland, OR
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Yifei Guo
- Department of Surgery, Oregon Health & Science University, Portland, OR
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Shaun M. Goodyear
- Division of Hematology and Oncology, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Jonathan R. Brody
- Department of Surgery, Oregon Health & Science University, Portland, OR
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| |
Collapse
|
6
|
Novoplansky O, Jagadeeshan S, Prasad M, Yegodayev KM, Marripati D, Shareb RA, Greenshpan Y, Mathukkada S, Ben-Lulu T, Bhattacharya B, Porgador A, Kong D, Brägelmann J, Gutkind JS, Elkabets M. Dual inhibition of HERs and PD-1 counteract resistance in KRAS G12C-mutant head and neck cancer. J Exp Clin Cancer Res 2024; 43:308. [PMID: 39567998 PMCID: PMC11577641 DOI: 10.1186/s13046-024-03227-0] [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: 04/14/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Basket clinical trials targeting the KRASG12C-mutation in solid tumors have shown initial promise, including in orphan KRASG12C head and neck cancer (HNC). However, development of resistance to KRASG12C-mutant-specific inhibitors (KRASG12Ci) remains a major obstacle. Here, we investigated the intrinsic (tumor-cell autonomus) and tumor-microenvironment (TME) mechanisms of resistance to the KRASG12Ci-MRTX849 and AMG510 in a unique syngenic murine KRASG12C-mutated HNC cell line. METHODS Western-blotting was used for protein abundance and activation, overexpression, and ligand activation studies to verify the intrinsic mechanism of resistance to KRASG12Ci in KRASG12C-mutated HNC cell line, 4NQO-L. In vitro KRASG12C-acquired-resistant cells were developed from 4NQO-L (4NQO-L-AcR). MRTX849/lapatinib combination efficacy, and CD8+ T-cells depletion, were assessed in C57BL/6 J mice and supplementation of anti-PD-1 (αPD-1) to MRTX849/lapatinib was also performed in 4NQO-L- KRASG12Ci-senisitve and 4NQO-L-AcR tumors. Immunohistochemistry (IHC) and Immunoflourescence (IF) analyses were performed to profile the TME and programmed death-ligand 1 (PD-L1) expression in tumors. RESULTS Activation and upregulation of EGFR and HER2/3 (pan-HERs) are the intrinsic mechanism of resistance to KRASG12Ci in 4NQO-L cells, and blocking pan-HERs signaling with lapatinib enhanced MRTX849 efficacy in vitro by inhibiting the MAPK and AKT/mTOR pathways. 4NQO-L-AcR upregulated the expression of pan-HERs, and lapatinib treatment re-sensitized 4NQO-L-AcR to MRTX849. In mice, MRTX849 showed a slight anti-tumor effect, but in combination with lapatinib a significant tumor growth delay was observed, but all tumors progressed over time. Histopathology analysis of the TME revealed infiltration of CD8+ T-cells after treatment combination, and these CD8+ T-cells play a key role in MRTX849/lapatinib efficacy. MRTX849/lapatinib treatment upregulated PD-L1 overexpression in both stromal and tumor cells, which presumably suppressed CD8+ T-cells and enabled immune escape and tumor progression. Supplementation of αPD-1 prolonged the progression-free survival of 4NQO-L-bearing mice treated with MRTX849/lapatinib. MRTX849/lapatinib treatment delayed tumor growth of 4NQO-L-AcR in mice; however, the percentages of CD8+ T-cells in 4NQO-L-AcR were low, and supplementation of MRTX849/lapatinib with αPD-1 did not improve the outcome. CONCLUSIONS Our study highlights the critical need for blocking both intrinsic and extrinsic mechanisms of resistance for the prolonged response and shows that such treatment is ineffective in KRASG12Ci-AcR tumors.
Collapse
Affiliation(s)
- Ofra Novoplansky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Manu Prasad
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ksenia M Yegodayev
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Divyasree Marripati
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Raghda Abu Shareb
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yariv Greenshpan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sooraj Mathukkada
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Talal Ben-Lulu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Baisali Bhattacharya
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dexin Kong
- School of Pharmaceutical Sciences, Tianjin Medical University, Tianjin, China
| | - Johannes Brägelmann
- Department of Translational Genomics, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany
- Mildred Scheel School of Oncology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, 50937, Cologne, Germany
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel.
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
7
|
Aguirre AJ, Stanger BZ, Maitra A. Hope on the Horizon: A Revolution in KRAS Inhibition Is Creating a New Treatment Paradigm for Patients with Pancreatic Cancer. Cancer Res 2024; 84:2950-2953. [PMID: 39279379 DOI: 10.1158/0008-5472.can-24-1926] [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: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/18/2024]
Abstract
KRAS is the most frequently altered oncogene in pancreatic ductal adenocarcinoma, in which the aberrantly activated RAS signaling pathway plays pleiotropic roles in tumor initiation and maintenance. Nearly four decades after the discovery of the RAS oncoprotein, a multitude of pharmacologic inhibitors are now available that directly target mutant KRAS. This In Focus commentary, published simultaneously with the 2024 AACR Special Conference on Pancreatic Cancer, summarizes the current state of this rapidly changing field, including preclinical data and emerging clinical trends with respect to therapeutic efficacy, mechanisms of resistance, and potential combinations to maximize clinical benefit from this promising class of therapies.
Collapse
Affiliation(s)
- Andrew J Aguirre
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ben Z Stanger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anirban Maitra
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, Texas
- Sheikh Ahmed Center for Pancreatic Cancer Research, UT MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
8
|
Fu Y, Tao J, Liu T, Liu Y, Qiu J, Su D, Wang R, Luo W, Cao Z, Weng G, Zhang T, Zhao Y. Unbiasedly decoding the tumor microenvironment with single-cell multiomics analysis in pancreatic cancer. Mol Cancer 2024; 23:140. [PMID: 38982491 PMCID: PMC11232163 DOI: 10.1186/s12943-024-02050-7] [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: 05/07/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis and limited therapeutic options. Research on the tumor microenvironment (TME) of PDAC has propelled the development of immunotherapeutic and targeted therapeutic strategies with a promising future. The emergence of single-cell sequencing and mass spectrometry technologies, coupled with spatial omics, has collectively revealed the heterogeneity of the TME from a multiomics perspective, outlined the development trajectories of cell lineages, and revealed important functions of previously underrated myeloid cells and tumor stroma cells. Concurrently, these findings necessitated more refined annotations of biological functions at the cell cluster or single-cell level. Precise identification of all cell clusters is urgently needed to determine whether they have been investigated adequately and to identify target cell clusters with antitumor potential, design compatible treatment strategies, and determine treatment resistance. Here, we summarize recent research on the PDAC TME at the single-cell multiomics level, with an unbiased focus on the functions and potential classification bases of every cellular component within the TME, and look forward to the prospects of integrating single-cell multiomics data and retrospectively reusing bulk sequencing data, hoping to provide new insights into the PDAC TME.
Collapse
Affiliation(s)
- Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guihu Weng
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
9
|
Li H, Xu L, Cao H, Wang T, Yang S, Tong Y, Wang L, Liu Q. Analysis on the pathogenesis and treatment progress of NRG1 fusion-positive non-small cell lung cancer. Front Oncol 2024; 14:1405380. [PMID: 38957319 PMCID: PMC11217482 DOI: 10.3389/fonc.2024.1405380] [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: 03/22/2024] [Accepted: 06/06/2024] [Indexed: 07/04/2024] Open
Abstract
Lung cancer persistently leads as the primary cause of morbidity and mortality among malignancies. A notable increase in the prevalence of lung adenocarcinoma has become evident in recent years. Although targeted therapies have shown in treating certain subsets of non-small cell lung cancers (NSCLC), a significant proportion of patients still face suboptimal therapeutic outcomes. Neuregulin-1 (NRG1), a critical member of the NRG gene family, initially drew interest due to its distribution within the nascent ventricular endocardium, showcasing an exclusive presence in the endocardium and myocardial microvessels. Recent research has highlighted NRG1's pivotal role in the genesis and progression across a spectrum of tumors, influencing molecular perturbations across various tumor-associated signaling pathways. This review provides a concise overview of NRG1, including its expression patterns, configuration, and fusion partners. Additionally, we explore the unique features and potential therapeutic strategies for NRG1 fusion-positive occurrences within the context of NSCLC.
Collapse
Affiliation(s)
- Hongyan Li
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| | - Lina Xu
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| | - Hongshun Cao
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| | - Tianyi Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| | - Siwen Yang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| | - Yixin Tong
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| | - Linlin Wang
- Department of Thoracic Surgery, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| | - Qiang Liu
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Shenyang Chest Hospital & Tenth People’s Hospital, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Yamashita K, Kumamoto Y. CAFs-Associated Genes (CAFGs) in Pancreatic Ductal Adenocarcinoma (PDAC) and Novel Therapeutic Strategy. Int J Mol Sci 2024; 25:6003. [PMID: 38892190 PMCID: PMC11172745 DOI: 10.3390/ijms25116003] [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: 04/11/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive cancer with striking fibrosis, and its mortality rate is ranked second across human cancers. Cancer-associated fibroblasts (CAFs) play a critical role in PDAC progression, and we reviewed the molecular understanding of PDAC CAFs and novel therapeutic potential at present. CAFs-associated genes (CAFGs) were tentatively classified into three categories by stroma specificity representing stroma/epithelia expression ratios (SE ratios). The recent classification using single cell transcriptome technology clarified that CAFs were composed of myofibroblasts (myCAFs), inflammatory CAFs (iCAFs), and other minor ones (e.g., POSTN-CAFs and antigen presenting CAFs, apCAFs). LRRC15 is a myCAFs marker, and myCAFs depletion by diphtheria toxin induces the rapid accumulation of cytotoxic T lymphocytes (CTLs) and therefore augment PDL1 antibody treatments. This finding proposes that myCAFs may be a critical regulator of tumor immunity in terms of PDAC progression. myCAFs are located in CAFs adjacent to tumor cells, while iCAFs marked by PDPN and/or COL14A1 are distant from tumor cells, where hypoxic and acidic environments being located in iCAFs putatively due to poor blood supply is consistent with HIF1A and GPR68 expressions. iCAFs may be shared with SASP (secretion-associated phenotypes) in senescent CAFs. myCAFs are classically characterized by CAFGs induced by TGFB1, while chemoresistant CAFs with SASP may dependent on IL6 expression and accompanied by STAT3 activation. Recently, it was found that the unique metabolism of CAFs can be targeted to prevent PDAC progression, where PDAC cells utilize glucose, whereas CAFs in turn utilize lactate, which may be epigenetically regulated, mediated by its target genes including CXCR4. In summary, CAFs have unique molecular characteristics, which have been rigorously clarified as novel therapeutic targets of PDAC progression.
Collapse
Affiliation(s)
- Keishi Yamashita
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kitasato 1-15-1, Minami-ku, Sagamihara 252-0374, Japan
| | - Yusuke Kumamoto
- Department of General-Pediatric-Hepatobiliary Pancreatic Surgery, Kitasato University School of Medicine, Sagamihara 252-0374, Japan;
| |
Collapse
|
11
|
Singhal A, Li BT, O'Reilly EM. Targeting KRAS in cancer. Nat Med 2024; 30:969-983. [PMID: 38637634 PMCID: PMC11845254 DOI: 10.1038/s41591-024-02903-0] [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: 01/04/2024] [Accepted: 03/04/2024] [Indexed: 04/20/2024]
Abstract
RAS family variants-most of which involve KRAS-are the most commonly occurring hotspot mutations in human cancers and are associated with a poor prognosis. For almost four decades, KRAS has been considered undruggable, in part due to its structure, which lacks small-molecule binding sites. But recent developments in bioengineering, organic chemistry and related fields have provided the infrastructure to make direct KRAS targeting possible. The first successes occurred with allele-specific targeting of KRAS p.Gly12Cys (G12C) in non-small cell lung cancer, resulting in regulatory approval of two agents-sotorasib and adagrasib. Inhibitors targeting other variants beyond G12C have shown preliminary antitumor activity in highly refractory malignancies such as pancreatic cancer. Herein, we outline RAS pathobiology with a focus on KRAS, illustrate therapeutic approaches across a variety of malignancies, including emphasis on the 'on' and 'off' switch allele-specific and 'pan' RAS inhibitors, and review immunotherapeutic and other key combination RAS targeting strategies. We summarize mechanistic understanding of de novo and acquired resistance, review combination approaches, emerging technologies and drug development paradigms and outline a blueprint for the future of KRAS therapeutics with anticipated profound clinical impact.
Collapse
Affiliation(s)
- Anupriya Singhal
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bob T Li
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Early Drug Development Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Eileen M O'Reilly
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|