1
|
Li F, Shen F. Metastatic pancreatic cancer with activating BRAF V600E mutations: A case report. World J Clin Cases 2025; 13:101665. [DOI: 10.12998/wjcc.v13.i16.101665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/06/2024] [Accepted: 01/11/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly malignant tumor that is resistant to chemotherapy, radiotherapy and immunotherapy. Combination chemotherapy regimens are the standard first-line regimens for metastatic disease, with a median survival < 12 months. Although recurrent genomic alterations such as the BRAF V600E mutation have been reported in PC, evidence supporting the clinical effectiveness of molecularly guided targeted therapies is limited.
CASE SUMMARY We report a case of a 33-year-old male who was referred to our department with weight loss of 5 kg in 2 months, anorexia and abdominal pain. Imaging showed extensive lesions involving the pancreas, liver, bones, muscles and lymph nodes accompanied by elevated carbohydrate antigen 19-9 (CA19-9) and carcinoembryonic antigen (CEA). Biopsy yielded a diagnosis of PC. Treatment with gemcitabine and nab-paclitaxel was initiated, but the disease progressed in < 2 months even though the patient’s general condition improved. Molecular testing revealed the presence of BRAF mutation. Dabrafenib/trametinib combination therapy was introduced, and the patient was treated for 2 months with a decrease in CA19-9 and CEA levels, but he died after 2 months of treatment.
CONCLUSION BRAF alterations are infrequent in PC. This case highlights the significance of molecular profiling in patients with PC, especially in patients with a high tumor burden.
Collapse
Affiliation(s)
- Fang Li
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, Fujian Province, China
- Xiamen Clinical Research Center for Cancer, Xiamen 361015, Fujian Province, China
- Clinical Research Center for Precision Medicine of Abdominal Tumor of Fujian Province, Xiamen 361015, Fujian Province, China
| | - Feng Shen
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, Fujian Province, China
- Xiamen Clinical Research Center for Cancer, Xiamen 361015, Fujian Province, China
- Clinical Research Center for Precision Medicine of Abdominal Tumor of Fujian Province, Xiamen 361015, Fujian Province, China
| |
Collapse
|
2
|
Subbiah V, Othus M, Palma J, Cuglievan B, Kurzrock R. Designing Clinical Trials for Patients With Rare Cancers: Connecting the Zebras. Am Soc Clin Oncol Educ Book 2025; 45:e100051. [PMID: 40228175 DOI: 10.1200/edbk-25-100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
The field of rare cancer research is rapidly transforming, marked by significant progress in clinical trials and treatment strategies. Rare cancers, as defined by the National Cancer Institute, occur in fewer than 150 cases per million people each year, yet they collectively represent a significant portion of all cancer diagnoses. Because of their infrequency, these cancers pose distinct challenges for clinical trials, including limited patient populations, geographical dispersion, and a general lack of awareness of treatment options. Economic limitations further complicate drug development, making initiatives such as the Orphan Drug Act essential for incentivizing research. The advent of next-generation sequencing (NGS) and precision medicine has been instrumental in identifying actionable genetic alterations in parallel with an explosion in the development of genomically targeted therapies, immunotherapies, and antibody drug conjugates. Advances in clinical NGS, precision medicine, and tumor-agnostic therapies have become central to the progress in rare cancer research. The development and approval of tumor-agnostic drugs, such as BRAF, NTRK, and RET inhibitors, and immunotherapy for mismatch repair deficient/microsatellite instability-high status cancers highlight the potential of personalized treatments across diverse cancer types and across the age spectrum. Collaborative trials from cooperative groups including SWOG DART, ASCO TAPUR, NCI-MATCH, pediatric COG-match, DRUP, IMPRESS, and innovative registrational basket and platform trials (eg, VE-Basket, ROAR, LIBRETTO-001, ARROW), along with patient advocacy group-run trials like TRACK, are enhancing access to clinical trials. In addition, artificial intelligence has the potential to improve the trial matching process. An integrated approach, combining these innovations in collaboration with multiple stakeholders, is crucial for advancing rare cancer research, offering hope for better patient outcomes and quality of life.
Collapse
Affiliation(s)
| | - Megan Othus
- SWOG Cancer Research Network Statistical Center, Seattle, WA
- Division of Public Health, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jim Palma
- TargetCancer Foundation, Rare Cancer Patient Advocacy Group, Cambridge, MA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, and Medical College of Wisconsin Cancer Center, Milwaukee, WI
- WIN Consortium, Paris, France
- University of Nebraska, Lincoln, NE
| |
Collapse
|
3
|
Janati-Fard F, Housaindokht MR, Moosavi F, Nakhaei-Rad S. Structural Insights Into the Impact of the Glycine-Rich Loop Mutation in Noonan Syndrome on the ATP Binding Pocket of CRAF Kinase. Proteins 2025; 93:1022-1034. [PMID: 39739408 DOI: 10.1002/prot.26769] [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: 08/06/2024] [Revised: 10/19/2024] [Accepted: 11/07/2024] [Indexed: 01/02/2025]
Abstract
The pathogenic G361A variant of CRAF, associated with increased intrinsic kinase activity in Noonan syndrome (NS), remains poorly understood in terms of its molecular and structural impact on kinase activity. To elucidate the mechanistic implications of the glycine to alanine substitution at residue 361 in CRAF, we employed molecular dynamics simulations. Our findings reveal that this mutation predominantly affects the ATP binding pocket and critical intermolecular interactions within the active cleft that favors the phosphate transfer reaction. Notably, our data highlight significant alterations in key interactions involving Lys470/Asp486 and ATP.Mg2+ in CRAFG361A that are absent in wild-type CRAF. Additionally, we identified a novel interaction mode between Lys431 and γ-phosphate in wild-type CRAF, a residue evolutionarily conserved in CRAFs but not in related kinases such as BRAF, ARAF, and KSR1/2. Furthermore, observed shifts in the αC-helix and G-loop relative to the wild-type correlate with an enlarged ATP-binding cavity in the mutant, reflecting structural adaptations due to these mutations. Overall, these structural insights underscore the elevated intrinsic kinase activity of the CRAFG361A variant and provide crucial mechanistic details that could inform the development of specific inhibitors targeting this variant.
Collapse
Affiliation(s)
- Fatemeh Janati-Fard
- Stem Cell Biology, and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Fatemeh Moosavi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Saeideh Nakhaei-Rad
- Stem Cell Biology, and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
4
|
Nikanjam M, Kato S, Allen T, Sicklick JK, Kurzrock R. Novel clinical trial designs emerging from the molecular reclassification of cancer. CA Cancer J Clin 2025; 75:243-267. [PMID: 39841128 PMCID: PMC12061631 DOI: 10.3322/caac.21880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Next-generation sequencing has revealed the disruptive reality that advanced/metastatic cancers have complex and individually distinct genomic landscapes, necessitating a rethinking of treatment strategies and clinical trial designs. Indeed, the molecular reclassification of cancer suggests that it is the molecular underpinnings of the disease, rather than the tissue of origin, that mostly drives outcomes. Consequently, oncology clinical trials have evolved from standard phase 1, 2, and 3 tissue-specific studies; to tissue-specific, biomarker-driven trials; to tissue-agnostic trials untethered from histology (all drug-centered designs); and, ultimately, to patient-centered, N-of-1 precision medicine studies in which each patient receives a personalized, biomarker-matched therapy/combination of drugs. Innovative technologies beyond genomics, including those that address transcriptomics, immunomics, proteomics, functional impact, epigenetic changes, and metabolomics, are enabling further refinement and customization of therapy. Decentralized studies have the potential to improve access to trials and precision medicine approaches for underserved minorities. Evaluation of real-world data, assessment of patient-reported outcomes, use of registry protocols, interrogation of exceptional responders, and exploitation of synthetic arms have all contributed to personalized therapeutic approaches. With greater than 1 × 1012 potential patterns of genomic alterations and greater than 4.5 million possible three-drug combinations, the deployment of artificial intelligence/machine learning may be necessary for the optimization of individual therapy and, in the near future, also may permit the discovery of new treatments in real time.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology‐OncologyUniversity of California San DiegoLa JollaCaliforniaUSA
- Moores Cancer CenterUniversity of California San Diego HealthLa JollaCaliforniaUSA
| | - Shumei Kato
- Division of Hematology‐OncologyUniversity of California San DiegoLa JollaCaliforniaUSA
- Moores Cancer CenterUniversity of California San Diego HealthLa JollaCaliforniaUSA
| | | | - Jason K. Sicklick
- Moores Cancer CenterUniversity of California San Diego HealthLa JollaCaliforniaUSA
- Division of Surgical OncologyDepartment of SurgeryUniversity of California San DiegoSan DiegoCaliforniaUSA
- Department of PharmacologyUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer CenterMilwaukeeWisconsinUSA
- Worldwide Innovative Networking in Personalized Cancer Medicine ConsortiumParisFrance
| |
Collapse
|
5
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [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: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
6
|
Reinhorn D, Moskovitz M, Tap WD, Li BT. Targeting HER2 in lung cancers: Evolving treatment landscape and drug development strategies. Cancer 2025; 131 Suppl 1:e35780. [PMID: 40171885 DOI: 10.1002/cncr.35780] [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: 09/24/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 04/04/2025]
Abstract
ERBB2 (HER2) alterations, including mutations, amplifications, and overexpression are emerging therapeutic targets in non-small cell lung cancer (NSCLC). Despite recent advancements, standard first-line therapy remains chemotherapy with or without immunotherapy. Several therapies targeting HER2 are under development and have been evaluated in clinical trials with inconsistent efficacy, including monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates. A major landmark was recently reached when trastuzumab deruxtecan (T-DXd), an antibody-drug conjugate, became the first Food and Drug Administration (FDA)-approved therapy for pretreated HER2-mutant NSCLC, following the promising efficacy demonstrated in the DESTINY-Lung trials. Furthermore, T-DXd has shown efficacy across various tumor types harboring HER2 alterations in the DESTINY-PanTumor trials, leading to its recent pan-tumor FDA approval for HER2-positive solid tumors, highlighting the potential of tumor-agnostic drug development strategies. In this review, the authors describe the different HER2 alterations and their clinical consequences, including their impact on prognosis and response to standard therapies. They provide an up-to-date overview of the current treatment landscape and add a comprehensive review of pivotal and ongoing clinical trials of HER2-targeted therapies, including tumor-agnostic drug development strategies.
Collapse
Affiliation(s)
- Daniel Reinhorn
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mor Moskovitz
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - William D Tap
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Bob T Li
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medicine, Cornell University, New York, New York, USA
| |
Collapse
|
7
|
Fatemi N, Mirbahari SN, Tierling S, Sanjabi F, Shahrivari S, AmeliMojarad M, Amelimojarad M, Mirzaei Rezaei M, Nobaveh P, Totonchi M, Nazemalhosseini Mojarad E. Emerging Frontiers in Colorectal Cancer Therapy: From Targeted Molecules to Immunomodulatory Breakthroughs and Cell-Based Approaches. Dig Dis Sci 2025; 70:919-942. [PMID: 39869166 PMCID: PMC11919954 DOI: 10.1007/s10620-024-08774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 11/20/2024] [Indexed: 01/28/2025]
Abstract
Colorectal cancer (CRC) is ranked as the second leading cause of cancer-related deaths globally, necessitating urgent advancements in therapeutic approaches. The emergence of groundbreaking therapies, including chimeric antigen receptor-T (CAR-T) cell therapies, oncolytic viruses, and immune checkpoint inhibitors, marks a transformative era in oncology. These innovative modalities, tailored to individual genetic and molecular profiles, hold the promise of significantly enhancing patient outcomes. This comprehensive review explores the latest clinical trials and advancements, encompassing targeted molecular therapies, immunomodulatory agents, and cell-based therapies. By evaluating the strengths, limitations, and potential synergies of these approaches, this research aims to reshape the treatment landscape and improve clinical outcomes for CRC patients, offering new found hope for those who have exhausted conventional options. The culmination of this work is anticipated to pave the way for transformative clinical trials, ushering in a new era of personalized and effective CRC therapy.
Collapse
Affiliation(s)
- Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Nasim Mirbahari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, Tehran, Iran
| | - Sascha Tierling
- Department of Genetics/Epigenetics, Faculty NT, Life Sciences, Saarland University, Saarbrücken, Germany
| | - Fatemeh Sanjabi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical, Tehran, Iran
| | - Shabnam Shahrivari
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical, Tehran, Iran
| | - Mandana AmeliMojarad
- Department of Biology, Faculty of Basic Science, Kharrazi University, Tehran, Iran
| | - Melika Amelimojarad
- Department of Biology, Faculty of Basic Science, Kharrazi University, Tehran, Iran
| | - Meygol Mirzaei Rezaei
- School of Advanced Sciences and Technology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Parsa Nobaveh
- School of Advanced Sciences and Technology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Mehdi Totonchi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, Tehran, Iran
| | - Ehsan Nazemalhosseini Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Yeman St, Chamran Expressway, P.O. Box 19857-17413, Tehran, Iran.
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
8
|
Mori S, Fujiwara-Tani R, Ogata R, Ohmori H, Fujii K, Luo Y, Sasaki T, Nishiguchi Y, Bhawal UK, Kishi S, Kuniyasu H. Anti-Cancer and Pro-Immune Effects of Lauric Acid on Colorectal Cancer Cells. Int J Mol Sci 2025; 26:1953. [PMID: 40076581 PMCID: PMC11901037 DOI: 10.3390/ijms26051953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Lauric acid (LAA) is a 12-carbon medium-chain fatty acid that reportedly has antitumor and muscle-protecting effects. However, the details of these antitumor effects remain unclear. Therefore, in this study, we investigated the mechanism underlying the antitumor effects of LAA in CT26 and HT29 colorectal cancer (CRC) cell lines. Our in vitro findings demonstrated that LAA suppressed CRC cell proliferation, induced mitochondrial oxidative stress (reactive oxygen species (ROS)), inhibited oxidative phosphorylation (OXPHOS), and induced apoptosis. Moreover, in vivo analysis of LAA showed a more pronounced antitumor effect in CT26 cells in a syngeneic mouse tumor model than in vitro; therefore, we further investigated its impact on host antitumor immunity. We observed that LAA increased the number of effector T cells in mouse tumors, while in vitro LAA activated mouse splenocytes (SplC) and promoted OXPHOS. In two-dimensional co-culture of SplC and CT26 cells, LAA induced cell death in cancer cells. In three-dimensional co-culture, LAA promoted SplC infiltration and suppressed the formation of tumor spheres. Thus, LAA may exert antitumor effects through increased ROS production in cancer cells and effector T cell activation via increased energy metabolism. These results suggest that LAA, when used in combination with existing anti-cancer drugs, is likely to exhibit sensitizing effects in terms of both antitumor and antitumor immune effects, and future clinical studies are anticipated.
Collapse
Grants
- 23K16621 Ministry of Education, Culture, Sports, Science and Technology
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 23K10481 Ministry of Education, Culture, Sports, Science and Technology
- 21K11223 Ministry of Education, Culture, Sports, Science and Technology
- 22K16497 Ministry of Education, Culture, Sports, Science and Technology
- 21K06926 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Shiori Mori
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka 573-1010, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Ruiko Ogata
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Hitoshi Ohmori
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Kiyomu Fujii
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Yi Luo
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Takamitsu Sasaki
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Ujjal Kumar Bhawal
- Research Institute of Oral Science, School of Dentistry at Matsudo, Nihon University, Matsudo 271-8587, Japan;
| | - Shingo Kishi
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
- Department of Pathological Diagnosis, Nozaki Tokushukai Hospital, Daito 574-0074, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| |
Collapse
|
9
|
Yao S, Sun L, Lu Y, Zhu X, Xu R, Yang T, Tang H, Guo P, Zhu T. Eliminating VEGFA+ tumor-associated neutrophils by antibody-drug conjugates boosts antitumor immunity and potentiates PD-1 immunotherapy in preclinical models of cervical cancer. Cell Death Dis 2025; 16:115. [PMID: 39971940 PMCID: PMC11840153 DOI: 10.1038/s41419-025-07402-9] [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: 08/28/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
Tumor-associated neutrophils (TANs) actively interact with antibody-drug conjugates (ADCs) within the tumor microenvironment (TME), though the detailed mechanisms governing their response to ADC treatment remain to be fully elucidated. Herein, we explored how ICAM1-targeted ADCs affect TAN dynamics in preclinical models of cervical cancer. We discovered that I-DXd, our in-house ADC targeting cervical cancer, effectively eliminates tumor cells through specific antigen recognition while concurrently depleting pro-tumor VEGFA + TANs via Fcγ receptor-mediated phagocytosis. This dual action promotes an immunologically favorable TME. Through comprehensive preclinical studies, we established a foundational understanding of the synergistic benefits of combining I-DXd treatment with PD-1 immune checkpoint inhibition, thereby opening new avenues for therapeutic intervention in advanced cervical cancer.
Collapse
Affiliation(s)
- Shili Yao
- School of Materials Science and Engineering, Faculty of Medicine, Tianjin University, Tianjin, China
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Lu Sun
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ye Lu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Xiu Zhu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Rui Xu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Institute of Molecular Medicine, Hangzhou Institute for Advanced Study (UCAS), Hangzhou, China
| | - Tong Yang
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Huarong Tang
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China.
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
- Department of Gynecological Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Peng Guo
- School of Materials Science and Engineering, Faculty of Medicine, Tianjin University, Tianjin, China.
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China.
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Tao Zhu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China.
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
10
|
Wang Z, Gao L, Jia Z, Liu L, Gu A, Liu Z, Zhu Q, Zuo Y, Yang M, Wang S, Ma J, Zhang J, Qiu S, Li Z, Wang J, Xiang D, Liu F, Shao R, Li Y, Li M, Wei W, Liu Y. Full-length transcriptome atlas of gallbladder cancer reveals trastuzumab resistance conferred by ERBB2 alternative splicing. Signal Transduct Target Ther 2025; 10:54. [PMID: 39948369 PMCID: PMC11825701 DOI: 10.1038/s41392-025-02150-w] [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: 09/08/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Aberrant RNA alternative splicing in cancer generates varied novel isoforms and protein variants that facilitate cancer progression. Here, we employed the advanced long-read full-length transcriptome sequencing on gallbladder normal tissues, tumors, and cell lines to establish a comprehensive full-length gallbladder transcriptomic atlas. It is of note that receptor tyrosine kinases were one of the most dynamic components with highly variable transcript, with Erb-B2 receptor tyrosine kinase 2 (ERBB2) as a prime representative. A novel transcript, designated ERBB2 i14e, was identified for encoding a novel functional protein, and its protein expression was elevated in gallbladder cancer and strongly associated with worse prognosis. With the regulation of splicing factors ESRP1/2, ERBB2 i14e was alternatively spliced from intron 14 and the encoded i14e peptide was proved to facilitate the interaction with ERBB3 and downstream signaling activation of AKT. ERBB2 i14e was inducible and its expression attenuated anti-ERBB2 treatment efficacy in tumor xenografts. Further studies with patient derived xenografts models validated that ERBB2 i14e blockage with antisense oligonucleotide enhanced the tumor sensitivity to trastuzumab and its drug conjugates. Overall, this study provides a gallbladder specific long-read transcriptome profile and discovers a novel mechanism of trastuzumab resistance, thus ultimately devising strategies to improve trastuzumab therapy.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Li Gao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ziheng Jia
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Liguo Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Ao Gu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Zhaonan Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Qin Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Yichen Zuo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingjie Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Shijia Wang
- Changzhou No.2 People Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Jiyao Ma
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Jingyun Zhang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Shimei Qiu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Zhizhen Li
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongxi Xiang
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Fatao Liu
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
| | - Rong Shao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjing Li
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China.
| | - Maolan Li
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China.
| | - Wu Wei
- Lingang Laboratory, Shanghai, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation (CSRCT-SHANGHAI), Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University school of Medicine, Shanghai, China.
- Department of General Surgery, Jiading Branch of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Wang L, Wen S, Zhu W, Zhang Z, Cheng Y. Response to trastuzumab deruxtecan and delayed immune-related events in a patient with metastatic HER2-positive NSCLC: a case report and literature review. Front Oncol 2025; 14:1469438. [PMID: 39896189 PMCID: PMC11782023 DOI: 10.3389/fonc.2024.1469438] [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: 07/23/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
Trastuzumab deruxtecan (DS-8201) is an antibody-drug conjugate (ADC) designed to target HER2 mutations. We reported a case study demonstrating a favorable response to DS-8201 in a patient with HER2 mutation-positive non-small cell lung cancer (NSCLC) who exhibited resistance to initial immunotherapy, along with delayed immune-related events of hypoadrenocorticism occurring five months after discontinuation of immune checkpoint inhibitors. After reviewing the relevant literature, we discussed the mechanism of ADC agents underlying their anti-tumor activity and the potential impact of DS-8201 on the tumor microenvironment. This case highlights the efficacy of DS-8201 in NSCLC, particularly in individuals who have failed first-line immunotherapy, and provide valuable insights for clinicians exploring innovative strategies for the management of patients with lung cancer.
Collapse
Affiliation(s)
- Lu Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shidi Wen
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenjia Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhiyang Zhang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Melosky B, Juergens RA, Banerji S, Sacher A, Wheatley-Price P, Snow S, Tsao MS, Leighl NB, Martins I, Cheema P, Liu G, Chu QSC. The continually evolving landscape of novel therapies in oncogene-driven advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359241308784. [PMID: 39776537 PMCID: PMC11705342 DOI: 10.1177/17588359241308784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly heterogeneous disease that is frequently associated with a host of known oncogenic alterations. Advances in molecular diagnostics and drug development have facilitated the targeting of novel alterations such that the majority of NSCLC patients have driver mutations that are now clinically actionable. The goal of this review is to gain insights into clinical research and development principles by summary, analysis, and discussion of data on agents targeting known alterations in oncogene-driven, advanced NSCLC beyond those in the epidermal growth factor receptor (EGFR) and the anaplastic lymphoma kinase (ALK). A search of published and presented literature was conducted to identify prospective trials and integrated analyses reporting outcomes for agents targeting driver gene alterations (except those in EGFR and ALK) in molecularly selected, advanced NSCLC. Clinical efficacy data were extracted from eligible reports and summarized in text and tables. Findings show that research into alteration-directed therapies in oncogene-driven, advanced NSCLC is an extremely active research field. Ongoing research focuses on the expansion of new agents targeting both previously identified targets (particularly hepatocyte growth factor receptor (MET), human epidermal growth factor receptor 2 (HER2), and Kirsten rat sarcoma viral oncogene homolog (KRAS)) as well as novel, potentially actionable targets (such as neuregulin-1 (NRG1) and phosphatidylinositol 3-kinase (PI3K)). The refinement of biomarker selection criteria and the development of more selective and potent agents are allowing for increasingly specific and effective therapies and the expansion of clinically actionable alterations. Clinical advances in this field have resulted in a large number of regulatory approvals over the last 3 years. Future developments should focus on the continued application of alteration therapy matching principles and the exploration of novel ways to target oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BC Cancer Agency—Vancouver, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Shantanu Banerji
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Parneet Cheema
- William Osler Health System, University of Toronto, Brampton, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Quincy S. C. Chu
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Qi P, Sun Y, Pang Y, Liu J, Cai X, Huang S, Xu Q, Wang Q, Zhou X. Diagnostic Utility of a 90-Gene Expression Assay (Canhelp-Origin) for Patients with Metastatic Cancer with an Unclear or Unknown Diagnosis. Mol Diagn Ther 2025; 29:81-89. [PMID: 39333459 DOI: 10.1007/s40291-024-00746-6] [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] [Accepted: 09/15/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Metastatic cancers with unclear or unknown origins pose significant challenges in diagnosis and management, frequently leading to suboptimal outcomes. Studies have demonstrated that a 90-gene expression assay is effective in predicting the primary origin and guiding the site-specific therapy to improve prognosis. This study aimed to evaluate the clinical effectiveness of a 90-gene expression assay in patients with unclear or unknown diagnoses. METHODS The study encompassed patients for whom a 90-gene expression assay was requested as part of standard care. Data on patient demographics, tumor characteristics, and clinical history were collected. The assay's performance was evaluated by comparing its predicted tumor type with the final histopathological diagnosis. RESULTS Among 303 cases analyzed, a 90-gene expression assay successfully identified a molecular-based tumor type for 295 (97.4%) patients. Comparison with histopathological diagnosis revealed an overall agreement of 88.5% (170/192). In patients with a single suspected primary site (n = 140), the assay confirmed the suspected diagnosis in 90.7% of cases. For those with a differential diagnosis (n = 52), the assay narrowed down the possibilities in 82.7% of cases. Moreover, in cases where the histopathology report indicated cancer of unknown primary (n = 103), the assay offered a molecular tumor type prediction with potential clinical significance. CONCLUSIONS This study demonstrates the significant impact of a 90-gene expression assay on diagnosis and potential treatment selection for difficult-to-diagnose patients, highlighting its clinical value as a standardized molecular approach to streamline further diagnostic testing for patients with metastatic cancer of unclear or unknown origin. Further prospective study is required to assess whether employing molecular diagnostic classifiers enhances clinical outcomes in these patients.
Collapse
Affiliation(s)
- Peng Qi
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Yifeng Sun
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Canhelp Genomics Research Center, Canhelp Genomics Co., Ltd., No.22 Xinyan Road, Hangzhou, 310000, People's Republic of China
| | - Yue Pang
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Jing Liu
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Xu Cai
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Shenglin Huang
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qinghua Xu
- Canhelp Genomics Research Center, Canhelp Genomics Co., Ltd., No.22 Xinyan Road, Hangzhou, 310000, People's Republic of China.
| | - Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China.
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
14
|
Viganò L, Zanuso V, Fiz F, Cerri L, Laino ME, Ammirabile A, Ragaini EM, Viganò S, Terracciano LM, Francone M, Ieva F, Di Tommaso L, Rimassa L. CT-based radiogenomics of intrahepatic cholangiocarcinoma. Dig Liver Dis 2025; 57:118-124. [PMID: 39003163 DOI: 10.1016/j.dld.2024.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is an aggressive disease with increasing incidence and its genetic alterations could be the target of systemic therapies. AIMS To elucidate if radiomics extracted from computed tomography (CT) may non-invasively predict ICC genetic alterations. METHODS All consecutive patients with a diagnosis of a mass-forming ICC (01/2016-06/2022) were considered. Inclusion criteria were availability of a high-quality contrast-enhanced CT and molecular profiling by NGS or FISH for FGFR2 fusion/rearrangement. The CT scan at diagnosis was considered. Genetic analyses were performed on surgical specimens (resectable patients) or biopsies (unresectable ones). The radiomic features were extracted using the LifeX software. Multivariate predictive models of the commonest genetic alterations were built. RESULTS In the 90 enrolled patients (58 NGS/32 FISH, median age 65 years), the most common genetic alterations were FGFR2 (20/90), IDH1 (10/58), and KRAS (9/58). At internal validation, the combined clinical-radiomic models achieved the best performance for the prediction of FGFR2 (AUC = 0.892) and IDH1 status (AUC = 0.819), outperforming the pure clinical and radiomic models. The radiomic model for predicting KRAS mutations achieved an AUC = 0.767 (vs. 0.660 of the clinical model) without further improvements with the addition of clinical features. CONCLUSIONS CT-based radiomics provides a reliable non-invasive prediction of ICC genetic status with a major impact on therapeutic strategies.
Collapse
Affiliation(s)
- Luca Viganò
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Hepatobiliary Unit, Department of Minimally Invasive General & Oncologic Surgery, Humanitas Gavazzeni University Hospital, Bergamo, Italy.
| | - Valentina Zanuso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesco Fiz
- Nuclear Medicine Unit, Department of Diagnostic Imaging, Ente Ospedaliero "Ospedali Galliera", Genoa, Italy; Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital, Tübingen, Germany
| | - Luca Cerri
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Angela Ammirabile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Radiology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Elisa Maria Ragaini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Samuele Viganò
- MOX laboratory, Department of Mathematics, Politecnico di Milano, Milan, Italy
| | - Luigi Maria Terracciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Pathology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Radiology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Ieva
- MOX laboratory, Department of Mathematics, Politecnico di Milano, Milan, Italy; CHDS - Center for Health Data Science, Human Technopole, Milan, Italy
| | - Luca Di Tommaso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Pathology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
15
|
Venturini J, Massaro G, Lavacchi D, Rossini D, Pillozzi S, Caliman E, Pellegrini E, Antonuzzo L. The emerging HER2 landscape in colorectal cancer: the key to unveil the future treatment algorithm? Crit Rev Oncol Hematol 2024; 204:104515. [PMID: 39304034 DOI: 10.1016/j.critrevonc.2024.104515] [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: 02/02/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Colorectal cancer (CRC) represents a global health threat, standing as the second leading cause of cancer-related death worldwide. Targeted therapies brought new hope for the metastatic stage, which historically bore a very poor prognosis. Human epidermal growth receptor 2 (HER2) overexpression concerns about 5 % of the metastatic CRC (mCRC) patients, including both gene amplifications and point mutations. Albeit its controversial prognostic role, preclinical and clinical data indicate HER2 as a negative predictive biomarker of response to anti-EGFR therapies. Tissue and plasma-based NGS testing, could permit a precise identification of this resistance mechanism both at baseline and during treatment, thus guiding decision-making. Furthermore, promising results come from completed and ongoing randomized trials, testing HER2 as an actionable target. In this review, we discuss the available evidence on HER2 targeting in advanced CRC, analyzing its possible future role in the treatment algorithm.
Collapse
Affiliation(s)
- Jacopo Venturini
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Giulia Massaro
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Rossini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Department of Health Science, University of Florence, Florence 50139, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Elisa Pellegrini
- Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy.
| |
Collapse
|
16
|
Mitiushkina NV, Tiurin VI, Anuskina AA, Bordovskaya NA, Nalivalkina EA, Terina DM, Berkut MV, Shestakova AD, Syomina MV, Kuligina ES, Togo AV, Imyanitov EN. Use of 3' Rapid Amplification of cDNA Ends (3' RACE)-Based Targeted RNA Sequencing for Profiling of Druggable Genetic Alterations in Urothelial Carcinomas. Int J Mol Sci 2024; 25:12126. [PMID: 39596194 PMCID: PMC11594887 DOI: 10.3390/ijms252212126] [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: 09/27/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Targeted treatment of advanced or metastatic urothelial carcinomas (UCs) requires the identification of druggable mutations. This study describes the development of a 3' Rapid Amplification of cDNA Ends (3' RACE)-based targeted RNA sequencing panel which accounts for the status of all genes relevant to UC treatment, namely, FGFR1-4, KRAS, NRAS, BRAF, ERBB2 (HER2), CD274 (PD-L1) and PIK3CA. FGFR2/3-activating point mutations or fusions were found in 54/233 (23.2%) tumors. FGFR3 rearrangements were identified in 11 patients, with eight of them being undetectable by commonly used PCR kits. In addition, one tumor contained a high-copy FGFR2 gene amplification accompanied by strong overexpression of the gene. Mutations in RAS/RAF genes were present in 30/233 (12.9%) UCs and were mutually exclusive with alterations affecting FGFR2/3 genes. On the contrary, activating events in the HER2 oncogene (point mutations and overexpression), as well as PIK3CA mutations, which were relatively common, occurred with similar frequencies in RAS/RAF- or FGFR2/3-positive vs. negative samples. High PD-L1 mRNA expression was associated with advanced disease stage and was not observed in tumors with increased HER2 mRNA expression or in UCs with evidence for FGFR2/3 activation. Three of the studied carcinomas had high-level microsatellite instability (MSI). Overall, more than half of the UCs had potentially druggable genetic alterations. The proposed NGS panel permits comprehensive and cost-efficient analysis of UC-specific molecular targets and may be considered in clinical routine.
Collapse
Affiliation(s)
- Natalia V. Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Vladislav I. Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Aleksandra A. Anuskina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Natalia A. Bordovskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Ekaterina A. Nalivalkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Darya M. Terina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Mariya V. Berkut
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Anna D. Shestakova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Maria V. Syomina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
| | - Ekaterina Sh. Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Alexandr V. Togo
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
- 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; (N.V.M.); (V.I.T.); (N.A.B.); (E.A.N.); (D.M.T.); (M.V.B.); (A.D.S.); (M.V.S.); (E.S.K.); (A.V.T.)
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
17
|
Di Stasi V, Contaldo A, Birtolo LI, Shahini E. Interplay of Cardiometabolic Syndrome and Biliary Tract Cancer: A Comprehensive Analysis with Gender-Specific Insights. Cancers (Basel) 2024; 16:3432. [PMID: 39410050 PMCID: PMC11476000 DOI: 10.3390/cancers16193432] [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: 08/24/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/20/2024] Open
Abstract
BTC overall incidence is globally increasing. CCA, including its subtypes, is a form of BTC. MetS, obesity, MASLD, and diabetes are all linked to CCA in interconnected ways. The link between obesity and CCA is less well-defined in Eastern countries as compared to Western. Although more research is needed to determine the relationship between MASLD and extrahepatic CCA (eCCA), MASLD may be a concurrent risk factor for intrahepatic CCA, particularly in populations with established or unidentified underlying liver disease. Interestingly, the risk of biliary tract cancer (BTC) seemed to be higher in patients with shorter diabetes durations who were not treated with insulin. Therefore, early detection and prevention of chronic liver disease, as well as additional intervention studies, will undoubtedly be required to determine whether improvements to MetS, weight loss, and diabetes therapy can reduce the risk and progression of BTC. However, further studies are needed to understand how reproductive hormones are involved in causing BTC and to develop consistent treatment for patients. Finally, it is critical to carefully assess the cardiological risk in BTC patients due to their increased intrinsic cardiovascular risk, putting them at risk for thrombotic complications, cardiovascular death, cardiac metastasis, and nonbacterial thrombotic endocarditis. This review aimed to provide an updated summary of the relation between the abovementioned cardio-metabolic conditions and BTC.
Collapse
Affiliation(s)
- Vincenza Di Stasi
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Antonella Contaldo
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Lucia Ilaria Birtolo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Umberto I Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| |
Collapse
|
18
|
Habringer S, Ihlow J, Kleo K, Klostermann A, Schmidt M, Chai L, Knödler M, Leyvraz S, Sigler C, Sinn B, Maschmeyer G, Jegodka Y, Benary M, Ott CE, Tinhofer I, Schäfer R, Möbs M, Keller U, Keilholz U, Rieke DT. A diagnostic challenge of KIT p.V559D and BRAF p.G469A mutations in a paragastric mass. Oncologist 2024; 29:908-912. [PMID: 38886160 PMCID: PMC11448896 DOI: 10.1093/oncolo/oyae137] [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: 03/21/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
A patient with gastrointestinal stroma tumor (GIST) and KIT p.V559D and BRAF p.G469A alterations was referred to our institutional molecular tumor board (MTB) to discuss therapeutic implications. The patient had been diagnosed with B-cell chronic lymphocytic leukemia (CLL) years prior to the MTB presentation. GIST had been diagnosed 1 month earlier. After structured clinical annotation of the molecular alterations and interdisciplinary discussion, we considered BRAF/KIT co-mutation unlikely in a treatment-naïve GIST. Discordant variant allele frequencies furthermore suggested a second malignancy. NGS of a CLL sample revealed the identical class 2 BRAF alteration, thus supporting admixture of CLL cells in the paragastric mass, leading to the detection of 2 alterations. Following the MTB recommendation, the patient received imatinib and had a radiographic response. Structured annotation and interdisciplinary discussion in specialized tumor boards facilitate the clinical management of complex molecular findings. Coexisting malignancies and clonal hematopoiesis warrant consideration in case of complex and uncommon molecular findings.
Collapse
Affiliation(s)
- Stefan Habringer
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 12203, Germany
- German Cancer Consortium (DKTK), Berlin 10115, Germany
| | - Jana Ihlow
- Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin 10178, Germany
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Karsten Kleo
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Anna Klostermann
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Max Schmidt
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Lidan Chai
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Maren Knödler
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Serge Leyvraz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Christian Sigler
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Bruno Sinn
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Georg Maschmeyer
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 12203, Germany
| | - Yvette Jegodka
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Manuela Benary
- Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin 10178, Germany
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Claus-Eric Ott
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin 10115, Germany
| | - Reinhold Schäfer
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Markus Möbs
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 12203, Germany
- German Cancer Consortium (DKTK), Berlin 10115, Germany
| | - Ulrich Keilholz
- German Cancer Consortium (DKTK), Berlin 10115, Germany
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Damian T Rieke
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 12203, Germany
- German Cancer Consortium (DKTK), Berlin 10115, Germany
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| |
Collapse
|
19
|
Kim TY, Kim SY, Kim JH, Jung HA, Choi YJ, Hwang IG, Cha Y, Lee GW, Lee YG, Kim TM, Lee SH, Lee S, Yun H, Choi YL, Yoon S, Han SW, Kim TY, Kim TW, Zang DY, Kang JH. Nationwide precision oncology pilot study: KOrean Precision Medicine Networking Group Study of MOlecular profiling-guided therapy based on genomic alterations in advanced solid tumors (KOSMOS) KCSG AL-20-05. ESMO Open 2024; 9:103709. [PMID: 39305545 PMCID: PMC11440300 DOI: 10.1016/j.esmoop.2024.103709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) has become widely available but molecular profiling-guided therapy (MGT) had not been well established in the real world due to lack of available therapies and expertise to match treatment. Our study was designed to test the feasibility of a nationwide platform of NGS-guided MGT recommended by a central molecular tumor board (cMTB) for metastatic solid tumors. PATIENTS AND METHODS Patients with advanced or metastatic solid tumors with available NGS results and without standard treatment were enrolled. The cMTB interpreted the patients' NGS reports and recommended the following: (i) investigational medicinal products (IMPs) approved in other indications; (ii) alternative treatments; (iii) clinical trials. The primary variables were the proportion of patients with actionable genomic alterations and those receiving MGT as per cMTB recommendations. Others included treatment duration (TD), overall response rate (ORR), disease control rate (DCR), and safety. RESULTS From February 2021 to February 2022, 193 cases [99 (51.3%) men; median age 58 years (range 24-88 years); median line of previous treatment 3 (range 0-9)] from 29 sites were enrolled for 60 cMTB sessions. The median time from case submission to cMTB discussion was 7 days (range 2-20 days), and to IMP treatment initiation was 28 days (range 14-90 days). Actionable genetic alterations were found in 145 patients (75.1%). A total of 89 (46.1%) patients received actual dosing of IMPs, and 10 (5.2%) were enrolled in cMTB-recommended clinical trials, achieving an MGT rate of 51.3%. ORR and DCR of IMPs were 10.1% and 72.5%, respectively. The median TD was 3.5 months [95% confidence interval (CI) 2.8-5.5 months], and the 4-month TD rate was 44.9%. The median overall survival of patients who received IMPs was 6.9 months (95% CI 5.2-10.0 months). CONCLUSION KOSMOS confirmed the feasibility of MGT recommended by the cMTB, achieving a high MGT match rate and promising effectiveness in heavily pretreated advanced cancer patients.
Collapse
Affiliation(s)
- T-Y Kim
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - S Y Kim
- Department of Oncology, University of Ulsan College of Medicine, Aan Medical Center, Seoul
| | - J H Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam.
| | - H A Jung
- Division of Hematology-Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul
| | - Y J Choi
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul
| | - I G Hwang
- Department of Internal Medicine, Chung-Ang University College of Medicine, Chung-Ang University Hospital, Seoul
| | - Y Cha
- Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Goyang
| | - G-W Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, Jinju
| | - Y-G Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Seoul
| | - T M Kim
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - S-H Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul
| | - S Lee
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul
| | - H Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul
| | - Y L Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| | - S Yoon
- Department of Oncology, University of Ulsan College of Medicine, Aan Medical Center, Seoul
| | - S W Han
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - T-Y Kim
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul
| | - T W Kim
- Department of Oncology, University of Ulsan College of Medicine, Aan Medical Center, Seoul
| | - D Y Zang
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University Medical Center, Anyang
| | - J H Kang
- Department of Medical Oncology, Seoul St. Mary's Hospital, The Catholic University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Parikh PM, Bahl A, Sharma G, Pramanik R, Wadhwa J, Bajpai P, Jandyal S, Dubey AP, Sarin A, Dadhich SC, Saklani AP, Kumar A, Chandra A, Rawat S, Selvasekar C, Aggarwal S. Management of Metastatic Colorectal Cancer (mCRC): Real-World Recommendations. South Asian J Cancer 2024; 13:287-295. [PMID: 40060353 PMCID: PMC11888815 DOI: 10.1055/s-0044-1791689] [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: 04/02/2025] Open
Abstract
INTRODUCTION Metastatic CRC is considered as a heterogenous disease. Its management is therefore complex and dynamic. In order the give a ready reference to community oncologists, we developed this real world recommendations. METHODS A group of experts with academic background and real world experience in mCRC got together. We reviewed the current literature and the insights gained from our real world experience. Based on the same we put together these recommendations. RECOMMENDATIONS RESULTS Molecular testing should be done wherever possible. Most of these patients will be treated with a palliative approach. Doublet chemotherapy is a long-standing standard of care. Triplet therapy may be offered where a more aggressive approach is indicated. Combination with anti -vascular endothelial growth factor antibodies and/or anti EGFR antibodies is also considered standard. In the first-line setting, pembrolizumab can be used for patients with mCRC and microsatellite instability-high or deficient mismatch repair tumours; Left and right sided tumours are distinct entities. Combination of chemotherapy and targeted therapy is used as per individual patient and tumour characteristics.Oligometastatic disease can be approached with potentially curative intent. Cytoreductive surgery plus chemotherapy can be offered to selected patients with peritoneal only metastases. Stereotactic body radiation therapy can be used as local therapy for patients with oligometastatic liver only disease who cannot be taken up for surgery. New strategies include induction-maintenance chemotherapy and perioperative chemotherapy. All drugs/ regimen included as standard of care in the first line can also be used in subsequent lines. Specific targetable driver mutation tumours can be treated accordingly with their complementary biological therapy. CONCLUSION Multidisciplinary team management and shared decision making are possible when patient and caregivers choose to become active participants.
Collapse
Affiliation(s)
- Purvish M. Parikh
- Department of Clinical Hematology, Sri Ram Cancer Center, Mahatma Gandhi University of Medical Sciences and Technology, Jaipur, Rajasthan, India
| | - Ankur Bahl
- Department of Medical Oncology, Fortis Hospital, Gurugram, Haryana, India
| | - Gopal Sharma
- Department of Medical Oncology, Max Healthcare Hospital, New Delhi, India
| | - Raja Pramanik
- Department of Medical Oncology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Jyoti Wadhwa
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Peush Bajpai
- Department of Medical Oncology, Manipal Hospital, New Delhi, India
| | - Sunny Jandyal
- Department of Medical Oncology, Action Cancer Hospital, New Delhi, India
| | - A P. Dubey
- Department of Medical Oncology, Delhi Heart and Lung Institute, New Delhi, India
| | - Aditya Sarin
- Department of Medical Oncology, Sir Ganga Ram Hospital, New Delhi, India
| | | | - Avinash P. Saklani
- Department of Surgical Gastroenterology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Ashok Kumar
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGI), Lucknow, Uttar Pradesh, India
| | - Abhijit Chandra
- Department of Surgical Gastroenterology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Saumitra Rawat
- Department of Surgical Gastroenterology, Sir Ganga Ram Hospital, New Delhi, India
| | - C. Selvasekar
- Clinical Services and Specialist Surgery, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Shyam Aggarwal
- Department of Medical Oncology, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
21
|
Kerioui M, Iasonos A, Gönen M, Arfé A. New clinical trial design borrowing information across patient subgroups based on fusion-penalized regression models. Stat Methods Med Res 2024; 33:1718-1730. [PMID: 39158499 DOI: 10.1177/09622802241267355] [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: 08/20/2024]
Abstract
In cancer research, basket trials aim to assess the efficacy of a drug using baskets, wherein patients are organized into subgroups according to their tumor type. In this context, using information borrowing strategy may increase the probability of detecting drug efficacy in active baskets, by shrinking together the estimates of the parameters characterizing the drug efficacy in baskets with similar drug activity. Here, we propose to use fusion-penalized logistic regression models to borrow information in the setting of a phase 2 single-arm basket trial with binary outcome. We describe our proposed strategy and assess its performance via a simulation study. We assessed the impact of heterogeneity in drug efficacy, prevalence of each tumor types and implementation of interim analyses on the operating characteristics of our proposed design. We compared our approach with two existing designs, relying on the specification of prior information in a Bayesian framework to borrow information across similar baskets. Notably, our approach performed well when the effect of the drug varied greatly across the baskets. Our approach offers several advantages, including limited implementation efforts and fast computation, which is essential when planning a new trial as such planning requires intensive simulation studies.
Collapse
Affiliation(s)
- Marion Kerioui
- Department of Biostatistics, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Alexia Iasonos
- Department of Biostatistics, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Mithat Gönen
- Department of Biostatistics, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Andrea Arfé
- Department of Biostatistics, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| |
Collapse
|
22
|
Lin C, Zhou KI, Green MF, Caughey BA, Strickler JH, Datto MB, McKinney MS. Comprehensive Genomic Profiling in Non-Myeloid Hematologic Malignancies Identifies Variants That Can Alter Clinical Practice. Hematol Rep 2024; 16:603-611. [PMID: 39449302 PMCID: PMC11503333 DOI: 10.3390/hematolrep16040059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Comprehensive genomic profiling (CGP) is frequently adopted to direct the clinical care of myeloid neoplasms and solid tumors, but its utility in the care of lymphoid and histiocytic cancers is less well defined. METHODS In this study, we aimed to evaluate the frequency at which mutations identified by CGP altered management in non-myeloid hematologic malignancies. We retrospectively examined the CGP results of 105 samples from 101 patients with non-myeloid hematologic malignancies treated at an academic medical center who had CGP testing between 2014 and 2021. RESULTS CGP revealed one or more pathogenic or likely pathogenic variant in 92 (88%) of samples and 73 (72%) of tested patients had one or more mutations with diagnostic, prognostic, or therapeutic significance. The identification of a resistance variant resulted in the suspension of the active treatment or affected subsequent treatment choice in 9 (69%) out of 13 patients. However, the presence of a therapy sensitizing variant only led to consideration of a biomarker-directed therapy in 6 (10%) out of 61 patients. CONCLUSIONS Overall, CGP of non-myeloid hematologic malignancies identified clinically significant variants in 72% of patients and resulted in a change in management in 22% of patients.
Collapse
Affiliation(s)
- Chenyu Lin
- Division of Hematologic Malignancies & Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC 27705, USA
| | - Katherine I. Zhou
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, NC 27705, USA
| | | | - Bennett A. Caughey
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - John H. Strickler
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, NC 27705, USA
| | - Michael B. Datto
- Department of Pathology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Matthew S. McKinney
- Division of Hematologic Malignancies & Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC 27705, USA
| |
Collapse
|
23
|
De S, Ehrlich M. Arrest and Attack: Microtubule-Targeting Agents and Oncolytic Viruses Employ Complementary Mechanisms to Enhance Anti-Tumor Therapy Efficacy. Genes (Basel) 2024; 15:1193. [PMID: 39336785 PMCID: PMC11431212 DOI: 10.3390/genes15091193] [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: 07/18/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Oncolytic viruses (OVs) are promising cancer immunotherapy agents that stimulate anti-tumor immunity through the preferential infection and killing of tumor cells. OVs are currently under limited clinical usage, due in part to their restricted efficacy as monotherapies. Current efforts for enhancement of the therapeutic potency of OVs involve their combination with other therapy modalities, aiming at the concomitant exploitation of complementary tumor weaknesses. In this context, microtubule-targeting agents (MTAs) pose as an enticing option, as they perturb microtubule dynamics and function, induce cell-cycle arrest, and cause mitotic cell death. MTAs induce therapeutic benefit through cancer-cell-autonomous and non-cell-autonomous mechanisms and are a main component of the standard of care for different malignancies. However, off-target effects and acquired resistance involving distinct cellular and molecular mechanisms may limit the overall efficacy of MTA-based therapy. When combined, OVs and MTAs may enhance therapeutic efficacy through increases in OV infection and immunogenic cell death and a decreased probability of acquired resistance. In this review, we introduce OVs and MTAs, describe molecular features of their activity in cancer cells, and discuss studies and clinical trials in which the combination has been tested.
Collapse
Affiliation(s)
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| |
Collapse
|
24
|
Elhariri A, Patel J, Mahadevia H, Albelal D, Ahmed AK, Jones JC, Borad MJ, Babiker H. Identifying Actionable Alterations in KRAS Wild-Type Pancreatic Cancer. Target Oncol 2024; 19:679-689. [PMID: 39123077 DOI: 10.1007/s11523-024-01088-3] [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: 07/22/2024] [Indexed: 08/12/2024]
Abstract
The 5-year relative survival rate for pancreatic cancer is currently the lowest among all cancer types with a dismal 13%. A Kirsten rat sarcoma virus (KRAS) gene mutation is present in approximately 90% of patients with pancreatic cancer; however, KRAS-specific drugs are not yet widely used in clinical practice for pancreatic cancer, specifically the KRASG12D variant. Advances in genomic testing revealed an opportunity to detect genetic alterations in a subset of patients with no KRAS mutation termed KRAS wild-type. Patients with KRAS wild-type tumors have a propensity to express driver alterations, hence paving the way for utilizing a targeted therapy approach either via clinical trials or standard-of-care drugs. These alterations include fusions, amplifications, translocations, rearrangements and microsatellite instability-high tumors and can be as high as 11% in some studies. Here, we discuss some of the most notable alterations in KRAS wild-type and highlight promising clinical trials.
Collapse
Affiliation(s)
- Ahmed Elhariri
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Jaydeepbhai Patel
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Himil Mahadevia
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Douaa Albelal
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Ahmed K Ahmed
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Jeremy C Jones
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Mitesh J Borad
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Hani Babiker
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| |
Collapse
|
25
|
Pu X, Li L, Xu F, Wang Z, Fu Y, Wu H, Ren J, Chen J, Sun B. HER2 amplification subtype intrahepatic cholangiocarcinoma exhibits high mutation burden and T cell exhaustion microenvironment. J Cancer Res Clin Oncol 2024; 150:403. [PMID: 39198311 PMCID: PMC11358322 DOI: 10.1007/s00432-024-05894-0] [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: 01/16/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024]
Abstract
OBJECTIVE This study aimed to establish a uniform standard for the interpretation of HER2 gene and protein statuses in intrahepatic cholangiocarcinoma (ICC). We also intended to explore the clinical pathological characteristics, molecular features, RNA expression and immune microenvironment of HER2-positive ICC. METHODS We analyzed a cohort of 304 ICCs using immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) to identify HER2 status. Comprehensive analyses of the clinicopathological, molecular genetic, and RNA expression characterizations of ICCs with varying HER2 statuses were performed using next-generation sequencing. We further investigated the tumor microenvironment of ICCs with different HER2 statuses using IHC and multiplex immunofluorescence staining. RESULTS HER2/CEP17 ratio of ≥ 2.0 and HER2 copy number ≥ 4.0; or HER2 copy number ≥ 6.0 were setup as FISH positive criteria. Based on this criterion, 13 (4.27%, 13/304) samples were classified as having HER2 amplification. The agreement between FISH and IHC results in ICC was poor. HER2-amplified cases demonstrated a higher tumor mutational burden compared to non-amplified cases. No significant differences were observed in immune markers between the two groups. However, an increased density of CD8 + CTLA4 + and CD8 + FOXP3 + cells was identified in HER2 gene-amplified cases. CONCLUSION FISH proves to be more appropriate as the gold standard for HER2 evaluation in ICC. HER2 gene-amplified ICCs exhibit poorer prognosis, higher mutational burden, and T cell exhaustion and immune suppressed microenvironment.
Collapse
Affiliation(s)
- Xiaohong Pu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Feng Xu
- Department of Medical Imaging, The Affiliated Suqian First People's Hospital of Nanjing Medical University, 223800, Suqian, Jiangsu Province, China
| | - Ziyu Wang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Hongyan Wu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Jun Ren
- Department of General Surgery, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China.
| | - Jun Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China.
| | - Beicheng Sun
- Medical School, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
26
|
Radić J, Nikolić I, Kolarov-Bjelobrk I, Vasiljević T, Djurić A, Vidović V, Kožik B. Prognostic and Predictive Significance of Primary Tumor Localization and HER2 Expression in the Treatment of Patients with KRAS Wild-Type Metastatic Colorectal Cancer: Single-Centre Experience from Serbia. J Pers Med 2024; 14:879. [PMID: 39202071 PMCID: PMC11355236 DOI: 10.3390/jpm14080879] [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: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
The treatment of patients with metastatic colorectal cancer (mCRC) is complex and is impacted by the location of the primary tumor (LPT). Our study aims to emphasize the importance of LPT as a prognostic and predictive marker as well as to examine the significance of HER2 overexpression in patients with mCRC, particularly in relation to the response to Epidermal Growth Factor Receptor Antibody treatment (anti-EGFR therapy). In this study, 181 patients with Kirsten RAS (KRAS) wild-type mCRC who received anti-EGFR therapy were included. Among them, 101 had left colon cancer (LCC) and 80 had right colon cancer (RCC). Results demonstrated that patients with KRAS wild-type LCC had better median overall survival (OS) (43 vs. 33 months, p = 0.005) and progression-free survival (PFS) (6 vs. 3 months, p < 0.001) compared to those with RCC. Multivariate analysis identified mucinous adenocarcinoma (p < 0.001), RCC location (p = 0.022), perineural invasion (p = 0.034), and tumors at the resection margin (p = 0.001) as independent predictors of OS, while mucinous adenocarcinoma (p = 0.001) and RCC location (p = 0.004) independently correlated with significantly shorter PFS. In addition, human epidermal growth factor receptor 2 (HER2) positive expression was significantly associated with worse PFS compared to HER2 negative results (p < 0.001). In conclusion, LPT is an important marker for predicting outcomes in the treatment of wild-type mCRC using anti-EGFR therapy, since patients with RCC have a statistically significantly shorter PFS and OS. Further investigation is needed to understand the role of HER2 overexpression in wild-type mCRC, as these patients also exhibit shorter survival.
Collapse
Affiliation(s)
- Jelena Radić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Ivan Nikolić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Ivana Kolarov-Bjelobrk
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Tijana Vasiljević
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Pathology and Laboratory Diagnostic, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Aleksandar Djurić
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Vladimir Vidović
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Bojana Kožik
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
27
|
Ashouri K, Wong A, Mittal P, Torres-Gonzalez L, Lo JH, Soni S, Algaze S, Khoukaz T, Zhang W, Yang Y, Millstein J, Lenz HJ, Battaglin F. Exploring Predictive and Prognostic Biomarkers in Colorectal Cancer: A Comprehensive Review. Cancers (Basel) 2024; 16:2796. [PMID: 39199569 PMCID: PMC11353018 DOI: 10.3390/cancers16162796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Colorectal cancer (CRC) remains the second leading cause of cancer-related mortality worldwide. While immune checkpoint inhibitors have significantly improved patient outcomes, their effectiveness is mostly limited to tumors with microsatellite instability (MSI-H/dMMR) or an increased tumor mutational burden, which comprise 10% of cases. Advancing personalized medicine in CRC hinges on identifying predictive biomarkers to guide treatment decisions. This comprehensive review examines established tissue markers such as KRAS and HER2, highlighting their roles in resistance to anti-EGFR agents and discussing advances in targeted therapies for these markers. Additionally, this review summarizes encouraging data on promising therapeutic targets and highlights the clinical utility of liquid biopsies. By synthesizing current evidence and identifying knowledge gaps, this review provides clinicians and researchers with a contemporary understanding of the biomarker landscape in CRC. Finally, the review examines future directions and challenges in translating promising biomarkers into clinical practice, with the goal of enhancing personalized medicine approaches for colorectal cancer patients.
Collapse
Affiliation(s)
- Karam Ashouri
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Alexandra Wong
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Pooja Mittal
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Lesly Torres-Gonzalez
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Jae Ho Lo
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Sandra Algaze
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Taline Khoukaz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Yan Yang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Joshua Millstein
- Department of Population and Public Health Sciences, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| |
Collapse
|
28
|
Tavano F, Latiano A, Palmieri O, Gioffreda D, Latiano T, Gentile A, Tardio M, Latiano TP, Gentile M, Terracciano F, Perri F. Duodenal Fluid Analysis as a Rewarding Approach to Detect Low-Abundance Mutations in Biliopancreatic Cancers. Int J Mol Sci 2024; 25:8436. [PMID: 39126005 PMCID: PMC11312909 DOI: 10.3390/ijms25158436] [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: 06/20/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Diagnosis of biliopancreatic cancers by the available serum tumor markers, imaging, and histopathological tissue specimen examination remains a challenge. Circulating cell-free DNA derived from matched pairs of secretin-stimulated duodenal fluid (DF) and plasma from 10 patients with biliopancreatic diseases and 8 control subjects was analyzed using AmpliSeq™ HD technology for Ion Torrent Next-Generation Sequencing to evaluate the potential of liquid biopsy with DF in biliopancreatic cancers. The median cfDNA concentration was greater in DF-derived than in plasma-derived samples. A total of 13 variants were detected: 11 vs. 1 were exclusive for DF relative to the plasma source, and 1 was shared between the two body fluids. According to the four-tier systems, 10 clinical tier-I-II (76.9%), 1 tier-III (7.7%), and 2 tier-IV (15.4%) variants were identified. Notably, the 11 tier-I-III variants were exclusively found in DF-derived cfDNA from five patients with biliopancreatic cancers, and were detected in seven genes (KRAS, TP53, BRAF, CDKN2A, RNF43, GNAS, and PIK3CA); 82% of the tier-I-III variants had a low abundance, with a VAF < 6%. The mutational profiling of DF seems to be a reliable and promising tool for identifying cancer-associated alterations in malignant cancers of the biliopancreatic tract.
Collapse
Affiliation(s)
- Francesca Tavano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Anna Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Orazio Palmieri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Domenica Gioffreda
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tiziana Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Annamaria Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Matteo Tardio
- Department of Surgery, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tiziana Pia Latiano
- Department of Oncology, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Marco Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Fulvia Terracciano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Francesco Perri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| |
Collapse
|
29
|
El-Sayed MM, Bianco JR, Li Y, Fabian Z. Tumor-Agnostic Therapy-The Final Step Forward in the Cure for Human Neoplasms? Cells 2024; 13:1071. [PMID: 38920700 PMCID: PMC11201516 DOI: 10.3390/cells13121071] [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: 05/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer accounted for 10 million deaths in 2020, nearly one in every six deaths annually. Despite advancements, the contemporary clinical management of human neoplasms faces a number of challenges. Surgical removal of tumor tissues is often not possible technically, while radiation and chemotherapy pose the risk of damaging healthy cells, tissues, and organs, presenting complex clinical challenges. These require a paradigm shift in developing new therapeutic modalities moving towards a more personalized and targeted approach. The tumor-agnostic philosophy, one of these new modalities, focuses on characteristic molecular signatures of transformed cells independently of their traditional histopathological classification. These include commonly occurring DNA aberrations in cancer cells, shared metabolic features of their homeostasis or immune evasion measures of the tumor tissues. The first dedicated, FDA-approved tumor-agnostic agent's profound progression-free survival of 78% in mismatch repair-deficient colorectal cancer paved the way for the accelerated FDA approvals of novel tumor-agnostic therapeutic compounds. Here, we review the historical background, current status, and future perspectives of this new era of clinical oncology.
Collapse
Affiliation(s)
| | | | | | - Zsolt Fabian
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (M.M.E.-S.); (J.R.B.); (Y.L.)
| |
Collapse
|
30
|
Martínez-Pérez J, Torrado C, Domínguez-Cejudo MA, Valladares-Ayerbes M. Targeted Treatment against Cancer Stem Cells in Colorectal Cancer. Int J Mol Sci 2024; 25:6220. [PMID: 38892410 PMCID: PMC11172446 DOI: 10.3390/ijms25116220] [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: 04/27/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
The cancer stem cell (SC) theory proposes that a population of SCs serves as the driving force behind fundamental tumor processes, including metastasis, recurrence, and resistance to therapy. The standard of care for patients with stage III and high-risk stage II colorectal cancer (CRC) includes surgery and adjuvant chemotherapy. Fluoropyrimidines and their combination with oxaliplatin increased the cure rates, being able to eradicate the occult metastatic SC in a fraction of patients. The treatment for unresectable metastatic CRC is based on chemotherapy, antibodies to VEGF and EGFR, and tyrosine-kinase inhibitors. Immunotherapy is used in MSI-H tumors. Currently used drugs target dividing cells and, while often effective at debulking tumor mass, these agents have largely failed to cure metastatic disease. SCs are generated either due to genetic and epigenetic alterations in stem/progenitor cells or to the dedifferentiation of somatic cells where diverse signaling pathways such as Wnt/β-catenin, Hedgehog, Notch, TGF-β/SMAD, PI3K/Akt/mTOR, NF-κB, JAK/STAT, DNA damage response, and Hippo-YAP play a key role. Anti-neoplastic treatments could be improved by elimination of SCs, becoming an attractive target for the design of novel agents. Here, we present a review of clinical trials assessing the efficacy of targeted treatment focusing on these pathways in CRC.
Collapse
Affiliation(s)
- Julia Martínez-Pérez
- Medical Oncology Department, Hospital Universitario Virgen del Rocio (HUVR), Avenida de Manuel Siurot s/n, 41013 Seville, Spain;
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocio (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Avenida de Manuel Siurot s/n, 41013 Seville, Spain;
| | - Carlos Torrado
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - María A. Domínguez-Cejudo
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocio (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Avenida de Manuel Siurot s/n, 41013 Seville, Spain;
| | - Manuel Valladares-Ayerbes
- Medical Oncology Department, Hospital Universitario Virgen del Rocio (HUVR), Avenida de Manuel Siurot s/n, 41013 Seville, Spain;
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocio (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Avenida de Manuel Siurot s/n, 41013 Seville, Spain;
| |
Collapse
|
31
|
Li BT, Meric-Bernstam F, Bardia A, Naito Y, Siena S, Aftimos P, Anderson I, Curigliano G, de Miguel M, Kalra M, Oh DY, Park JO, Postel-Vinay S, Rha SY, Satoh T, Spanggaard I, Michelini F, Smith A, Machado KK, Saura C. Trastuzumab deruxtecan in patients with solid tumours harbouring specific activating HER2 mutations (DESTINY-PanTumor01): an international, phase 2 study. Lancet Oncol 2024; 25:707-719. [PMID: 38710187 DOI: 10.1016/s1470-2045(24)00140-2] [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: 11/14/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Trastuzumab deruxtecan is a HER2-directed antibody-drug conjugate approved by the US Food and Drug Administration and the European Medicines Agency for HER2-mutant non-small-cell lung cancer. Few treatment options exist for patients with HER2-mutant solid tumours beyond lung cancers. We investigated trastuzumab deruxtecan in metastatic solid tumours with specific activating HER2 mutations. METHODS In this open-label, phase 2, basket study done in 29 centres in Asia, Europe, and North America, we investigated trastuzumab deruxtecan (5·4 mg/kg every 3 weeks by intravenous infusion) in patients aged 18 years or older with unresectable or metastatic solid tumours with specific activating HER2 mutations, an Eastern Cooperative Oncology Group performance status of 0 or 1, and disease progression following previous treatment (previous HER2-targeted therapy was permitted) or with no satisfactory alternative treatment options. The primary endpoint was confirmed objective response rate by independent central review. Anti-tumour activity and safety were analysed in all patients who received at least one dose of trastuzumab deruxtecan. This trial is registered with ClinicalTrials.gov, NCT04639219, and is active but no longer recruiting. FINDINGS Between Dec 30, 2020, and Jan 25, 2023, 102 patients (62 [61%] female and 40 [39%] male; median age 66·5 years [IQR 58-72]; 51 [50%] White, two [2%] Black or African American, 38 [37%] Asian, and 11 [11%] did not have race information reported) with solid tumours with activating HER2 mutations received trastuzumab deruxtecan and were included in the anti-tumour activity and safety analyses sets. Patients had a median of three (IQR 2-4) previous treatment regimens. The median duration of follow-up was 8·61 months (IQR 3·71-12·68). The objective response rate by independent central review was 29·4% (95% CI 20·8-39·3; 30 of 102 patients). 52 (51%) patients had a treatment-emergent adverse event of grade 3 or worse; the most common events (in ≥5% of patients) were anaemia (16 [16%]) and neutrophil count decreased (eight [8%]). Drug-related treatment-emergent serious adverse events occurred in ten (10%) patients. Adjudicated drug-related interstitial lung disease or pneumonitis of any grade occurred in 11 patients (11%; three grade 1, five grade 2, one grade 3, and two grade 5); there were two (2%) cases of fatal adjudicated drug-related interstitial lung disease or pneumonitis. INTERPRETATION Trastuzumab deruxtecan showed anti-tumour activity and durable responses in heavily pretreated patients across multiple tumour types with activating HER2 mutations, with no new safety signals. Prespecified HER2 mutations might be targeted by HER2-directed antibody-drug conjugates and our findings support further investigation of trastuzumab deruxtecan in the pan-tumour setting. FUNDING AstraZeneca and Daiichi Sankyo.
Collapse
Affiliation(s)
- Bob T Li
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - Philippe Aftimos
- Institut Jules Bordet-Université Libre de Bruxelles, Brussels, Belgium
| | | | - Giuseppe Curigliano
- Istituto Europeo di Oncologia, IRCCS Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - Maria de Miguel
- START Madrid Centro Integral Oncológico Clara Campal (CIOCC)-HM Sanchinarro University Hospital, Madrid, Spain
| | - Maitri Kalra
- Ball Memorial Hospital (IU Health), Muncie, IN, USA
| | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea
| | - Joon Oh Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sophie Postel-Vinay
- Inserm Unit U981 Gustave Roussy, Villejuif, France; Cancer Institute, University College London, London, UK
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea
| | | | - Iben Spanggaard
- Department of Oncology, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark
| | - Flavia Michelini
- Translational Medicine, Oncology Research and Development, AstraZeneca, Waltham, MA, USA
| | - Ann Smith
- Oncology Biometrics, Oncology Research and Development, AstraZeneca, Cambridge, UK
| | - Karime Kalil Machado
- Late Development Oncology, Oncology Research and Development, AstraZeneca, Gaithersburg, MD, USA
| | - Cristina Saura
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
32
|
Murciano-Goroff YR, Uppal M, Chen M, Harada G, Schram AM. Basket Trials: Past, Present, and Future. ANNUAL REVIEW OF CANCER BIOLOGY 2024; 8:59-80. [PMID: 38938274 PMCID: PMC11210107 DOI: 10.1146/annurev-cancerbio-061421-012927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Large-scale tumor molecular profiling has revealed that diverse cancer histologies are driven by common pathways with unifying biomarkers that can be exploited therapeutically. Disease-agnostic basket trials have been increasingly utilized to test biomarker-driven therapies across cancer types. These trials have led to drug approvals and improved the lives of patients while simultaneously advancing our understanding of cancer biology. This review focuses on the practicalities of implementing basket trials, with an emphasis on molecularly targeted trials. We examine the biologic subtleties of genomic biomarker and patient selection, discuss previous successes in drug development facilitated by basket trials, describe certain novel targets and drugs, and emphasize practical considerations for participant recruitment and study design. This review also highlights strategies for aiding patient access to basket trials. As basket trials become more common, steps to ensure equitable implementation of these studies will be critical for molecularly targeted drug development.
Collapse
Affiliation(s)
| | - Manik Uppal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Monica Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Guilherme Harada
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
33
|
Abedizadeh R, Majidi F, Khorasani HR, Abedi H, Sabour D. Colorectal cancer: a comprehensive review of carcinogenesis, diagnosis, and novel strategies for classified treatments. Cancer Metastasis Rev 2024; 43:729-753. [PMID: 38112903 DOI: 10.1007/s10555-023-10158-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Colorectal cancer is the third most common and the second deadliest cancer worldwide. To date, colorectal cancer becomes one of the most important challenges of the health system in many countries. Since the clinical symptoms of this cancer appear in the final stages of the disease and there is a significant golden time between the formation of polyps and the onset of cancer, early diagnosis can play a significant role in reducing mortality. Today, in addition to colonoscopy, minimally invasive methods such as liquid biopsy have received much attention. The treatment of this complex disease has been mostly based on traditional treatments including surgery, radiotherapy, and chemotherapy; the high mortality rate indicates a lack of success for current treatment methods. Moreover, disease recurrence is another problem of traditional treatments. Recently, new approaches such as targeted therapy, immunotherapy, and nanomedicine have opened new doors for cancer treatment, some of which have already entered the market, and many methods have shown promising results in clinical trials. The success of immunotherapy in the treatment of refractory disease, the introduction of these methods into neoadjuvant therapy, and the successful results in tumor shrinkage without surgery have made immunotherapy a tough competitor for conventional treatments. It seems that the combination of those methods with such targeted therapies will go through promising changes in the future of colorectal cancer treatment.
Collapse
Affiliation(s)
- Roya Abedizadeh
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Fateme Majidi
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Hamid Reza Khorasani
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Hassan Abedi
- Department of Internal Medicine, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Davood Sabour
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran.
| |
Collapse
|
34
|
Devalle S, Aran V, Bastos Júnior CDS, Pannain VL, Brackmann P, Gregório ML, Ferreira Manso JE, Moura Neto V. A panorama of colon cancer in the era of liquid biopsy. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100148. [PMID: 40027146 PMCID: PMC11863817 DOI: 10.1016/j.jlb.2024.100148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 03/05/2025]
Abstract
Colon cancer (CC) is one of the most frequent cancers worldwide being responsible for over 500 thousand deaths in 2022. Its financial and human burden is expected to increase in the next decades accompanying the growing and aging of the global population. Much of this burden could be alleviated considering that the lethality of CC is mostly due to its late diagnosis and failure in the individualized management of patients. Coordinated government actions and implementation of better diagnostic tools capable of detecting CC earlier and of tracking tumoral evolution are mandatory to achieve a reduction in CC's social impact. CtDNA-based liquid biopsy (LB) has great potential to contribute to patients' screening adhesion, CC earlier detection, and to longitudinal tumor follow-up. In this review, we will discuss the latest epidemiological data on CC disease, diagnostic, subtypes, genetics, and treatment management focusing on the advantages and limitations of ctDNA-based LB, including important bottlenecks and solutions necessary for its clinical translation. The latest ctDNA-directed CC clinical trials will also be examined.
Collapse
Affiliation(s)
- Sylvie Devalle
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Veronica Aran
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Vera Lucia Pannain
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Brackmann
- Clínica de Coloproctologia do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - Marcelo Leal Gregório
- Instituto de Pesquisas Biomédicas do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - José Eduardo Ferreira Manso
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Dong F. Pan-Cancer Molecular Biomarkers: A Paradigm Shift in Diagnostic Pathology. Clin Lab Med 2024; 44:325-337. [PMID: 38821647 DOI: 10.1016/j.cll.2023.08.013] [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: 06/02/2024]
Abstract
The rapid adoption of next-generation sequencing in clinical oncology has enabled the detection of molecular biomarkers shared between multiple tumor types. These pan-cancer biomarkers include sequence-altering mutations, copy number changes, gene rearrangements, and mutational signatures and have been demonstrated to predict response to targeted therapy. This article reviews issues surrounding current and emerging pan-cancer molecular biomarkers in clinical oncology: technological advances that enable the broad detection of cancer mutations across hundreds of genes, the spectrum of driver and passenger mutations derived from human cancer genomes, and implications for patient care now and in the near future.
Collapse
Affiliation(s)
- Fei Dong
- Department of Pathology, Stanford University School of Medicine, 3375 Hillview Ave, Palo Alto, CA 94304, USA.
| |
Collapse
|
36
|
Puco K, Fagereng GL, Brabrand S, Niehusmann P, Støre Blix E, Samdal Steinskog ES, Haug Å, Fredvik Torkildsen C, Oppedal IA, Meltzer S, Flobak Å, Johansson KAM, Bjørge L, Hjortland GO, Dalhaug A, Lund JÅ, Gilje B, Grønlie Cameron M, Hovland R, Falk RS, Smeland S, Giercksky Russnes HE, Taskén K, Helland Å. IMPRESS-Norway: improving public cancer care by implementing precision medicine in Norway; inclusion rates and preliminary results. Acta Oncol 2024; 63:379-384. [PMID: 38779911 PMCID: PMC11332498 DOI: 10.2340/1651-226x.2024.28322] [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: 12/21/2023] [Accepted: 03/16/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND PURPOSE In Norway, comprehensive molecular tumour profiling is implemented as part of the public healthcare system. A substantial number of tumours harbour potentially targetable molecular alterations. Therapy outcomes may improve if targeted treatments are matched with actionable genomic alterations. In the IMPRESS-Norway trial (NCT04817956), patients are treated with drugs outside the labelled indication based on their tumours molecular profile. PATIENTS AND METHODS IMPRESS-Norway is a national, prospective, non-randomised, precision cancer medicine trial, offering treatment to patients with advanced-stage disease, progressing on standard treatment. Comprehensive next-generation sequencing, TruSight Oncology 500, is used for screening. Patients with tumours harbouring molecular alterations with matched targeted therapies available in IMPRESS-Norway, are offered treatment. Currently, 24 drugs are available in the study. Primary study endpoints are percentage of patients offered treatment in the trial, and disease control rate (DCR) defined as complete or partial response or stable disease in evaluable patients at 16 weeks (W16) of treatment. Secondary endpoint presented is DCR in all treated patients. RESULTS Between April 2021 and October 2023, 1,167 patients were screened, and an actionable mutation with matching drug was identified for 358 patients. By the data cut off 186 patients have initiated treatment, 170 had a minimum follow-up time of 16 weeks, and 145 also had evaluable disease. In patients with evaluable disease, the DCR was 40% (58/145). Secondary endpoint analysis of DCR in all treated patients, showed DCR of 34% (58/170). INTERPRETATION Precision cancer medicine demonstrates encouraging clinical effect in a subset of patients included in the IMPRESS-Norway trial.
Collapse
Affiliation(s)
- Katarina Puco
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Gro Live Fagereng
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | | | - Pitt Niehusmann
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Egil Støre Blix
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway
| | | | - Åse Haug
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | | | - Irja Alida Oppedal
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Sebastian Meltzer
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Åsmund Flobak
- Department of Oncology, Trondheim University Hospital, Trondheim, Norway
| | | | - Line Bjørge
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | | | | | - Jo-Åsmund Lund
- Clinic for Cancer Treatment and Rehabilitation, Møre and Romsdal Hospital Trust, Ålesund, Norway
| | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | | | - Randi Hovland
- Department of Cancer Genomics, Haukeland University Hospital, Bergen, Norway
| | - Ragnhild S Falk
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Sigbjørn Smeland
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Hege Elisabeth Giercksky Russnes
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Kjetil Taskén
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Åslaug Helland
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Oncology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
37
|
Zanuso V, Nash T, Casolino R, Armstrong G, Pallise O, Milne J, Braconi C. Insights for clinical management from the real-life data of the centralized West of Scotland biliary cancer clinic. BMC Cancer 2024; 24:597. [PMID: 38755562 PMCID: PMC11097428 DOI: 10.1186/s12885-024-12279-6] [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: 01/22/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND With the increasing of novel therapeutics for the treatment of Biliary Tract Cancers (BTC), and the need to assess their socio-economic impacts for national licence approvals, it is as important as ever to have real-life data in national populations. METHODS AND RESULTS We performed an audit of the first 2 year-activity (Sep 2019-Sep 2021) of the centralized West-of-Scotland-BTC clinic. 122 patients accessed the service, including 68% with cholangiocarcinoma (CCA), 27% with gallbladder cancer (GBC), and 5% with ampulla of Vater carcinoma with biliary phenotype (AVC). Median age at diagnosis was 66 (28-84), with 30% of newly diagnosed patients being younger than 60 years-old. Thirty-five cases (29%) underwent surgery, followed by adjuvant-chemotherapy in 66%. 60% had recurrent disease (80% with distant relapse). Sixty-four patients (58%) started first-line Systemic-AntiCancer-Treatment (SACT). Of these, 37% received second line SACT, the majority of which had iCCA and GBC. Thirty-% of those who progressed received third line SACT. CONCLUSIONS About 30% of BTC were eligible for curative surgery. Fifty-eight and twenty% of the overall cohort of advanced BTC patients received first and second line SACT. Our data suggest that reflex genomic profiling may not be cost-effective until molecularly driven strategies are limited to second line setting.
Collapse
Affiliation(s)
- Valentina Zanuso
- School of Cancer Sciences, University of Glasgow, G61 1QH, Glasgow, UK
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
| | - Tamsin Nash
- Beatson West of Scotland Cancer Centre, G12 0YN, Glasgow, UK
- NHS Greater Glasgow and Clyde, Glasgow, UK
| | | | | | - Ona Pallise
- Beatson West of Scotland Cancer Centre, G12 0YN, Glasgow, UK
- NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Jen Milne
- Beatson West of Scotland Cancer Centre, G12 0YN, Glasgow, UK
- NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Chiara Braconi
- School of Cancer Sciences, University of Glasgow, G61 1QH, Glasgow, UK.
- Beatson West of Scotland Cancer Centre, G12 0YN, Glasgow, UK.
- CRUK-Scotland Cancer Centre, Glasgow-Edinburgh, UK.
| |
Collapse
|
38
|
Gou Q, Gou Q, Gan X, Xie Y. Novel therapeutic strategies for rare mutations in non-small cell lung cancer. Sci Rep 2024; 14:10317. [PMID: 38705930 PMCID: PMC11070427 DOI: 10.1038/s41598-024-61087-2] [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: 12/27/2023] [Accepted: 04/30/2024] [Indexed: 05/07/2024] Open
Abstract
Lung cancer is still the leading cause of cancer-related mortality. Over the past two decades, the management of non-small cell lung cancer (NSCLC) has undergone a significant revolution. Since the first identification of activating mutations in the epidermal growth factor receptor (EGFR) gene in 2004, several genetic aberrations, such as anaplastic lymphoma kinase rearrangements (ALK), neurotrophic tropomyosin receptor kinase (NTRK) and hepatocyte growth factor receptor (MET), have been found. With the development of gene sequencing technology, the development of targeted drugs for rare mutations, such as multikinase inhibitors, has provided new strategies for treating lung cancer patients with rare mutations. Patients who harbor this type of oncologic driver might acquire a greater survival benefit from the use of targeted therapy than from the use of chemotherapy and immunotherapy. To date, more new agents and regimens can achieve satisfactory results in patients with NSCLC. In this review, we focus on recent advances and highlight the new approval of molecular targeted therapy for NSCLC patients with rare oncologic drivers.
Collapse
Affiliation(s)
- Qitao Gou
- Department of Radiation Oncology and Department of Head & Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiheng Gou
- Department of Radiation Oncology and Department of Head & Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiaochuan Gan
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxin Xie
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Huang Y, Zhao Y, Huang Y, Yang Y, Zhang Y, Hong S, Zhao H, Zhao S, Zhou T, Chen G, Zhou H, Ma Y, Zhou N, Zhang L, Fang W. Phase 1b trial of anti-HER2 antibody inetetamab and pan-HER inhibitor pyrotinib in HER2-positive advanced lung cancer. MedComm (Beijing) 2024; 5:e536. [PMID: 38685972 PMCID: PMC11057420 DOI: 10.1002/mco2.536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
There remains an unmet need for targeted therapies against advanced non-small-cell lung cancer (NSCLC) with HER2 mutations. To improve the antitumor activity of single anti-HER2 agent, this prospective, single-arm clinical trial (NCT05016544) examined the safety profile and efficacy of anti-HER2 antibody inetetamab and pan-HER TKI pyrotinib in HER2-posivite advanced NSCLC patients. Enrolled patients received inetetamab every 3 weeks and pyrotinib once per day (pyrotinib, dose-escalation part, 240 mg, 320 mg; dose-expansion part, 320 mg). Primary endpoints were dose-limiting toxicity (DLT) dosage and safety. Secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). A total of 48 patients were enrolled. During the dose-escalation period, no DLT occurred. Diarrhea was the most commonly reported treatment-related adverse event (TRAE). Grade 3 TRAEs occurred in seven patients. The median PFS (mPFS) was 5.5 months [95% confidence interval (CI): 4.4-8.6 months]. The confirmed ORR and DCR reached 25% (11/44) and 84.1% (37/44), respectively. Responses were shown in patients with distinct HER2 aberrations. In summary, inetetamab in combination with pyrotinib demonstrated acceptable safety and antitumor activity among patients with advanced HER2-mutant NSCLC.
Collapse
Affiliation(s)
- Yihua Huang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yuanyuan Zhao
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yan Huang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yunpeng Yang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yaxiong Zhang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Shaodong Hong
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Hongyun Zhao
- Department of Clinical ResearchState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Shen Zhao
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Ting Zhou
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Gang Chen
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Huaqiang Zhou
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yuxiang Ma
- Department of Clinical ResearchState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Ningning Zhou
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Li Zhang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Wenfeng Fang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| |
Collapse
|
40
|
Chen N, He L, Zou Q, Deng H. HER2 targeted therapy in colorectal Cancer: Current landscape and future directions. Biochem Pharmacol 2024; 223:116101. [PMID: 38442793 DOI: 10.1016/j.bcp.2024.116101] [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: 08/31/2023] [Revised: 02/11/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024]
Abstract
Colorectal cancer (CRC) is one of the most common causes of tumor-related deaths globally. Despite recent improvements in the comprehensive therapy of malignancy, metastatic CRC continues to have a poor prognosis. Human epidermal growth factor receptor 2 (HER2) is an established oncogenic driver, which is successfully targeted for breast and gastric cancers. Approximately 5% of CRC patients carry somatic HER2 mutations or gene amplification. In 2019, the U.S. Food and Drug Administration have approved trastuzumab and pertuzumab in combination with chemotherapy for the treatment of HER2-positive metastatic CRC. This approval marked a significant milestone in the treatment of CRC, as HER2-positive patients now have access to targeted therapies that can improve their outcomes. Yet, assessment for HER2 overexpression/ amplification in CRC has not been standardized. The resistance mechanisms to anti-HER2 therapy have been not clearly investigated in CRC. Although many unknowns remain, an improved understanding of these anti-HER2 agents will be essential for advanced CRC. In this review, we provide an overview of the role of HER2 in CRC as an oncogenic driver, a prognostic and predictive biomarker, and a clinically actionable target, as well as the current progress and challenges in the field.
Collapse
Affiliation(s)
- Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, China; Center of Science and Research, Chengdu Medical College, Chengdu, 610500, China
| | - Ling He
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qiang Zou
- Center of Science and Research, Chengdu Medical College, Chengdu, 610500, China.
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
41
|
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 PMCID: PMC11849277 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
|
42
|
DiPeri TP, Evans KW, Wang B, Zhao M, Akcakanat A, Raso MG, Rizvi YQ, Zheng X, Korkut A, Varadarajan K, Uzunparmak B, Dumbrava EE, Pant S, Ajani JA, Pohlmann PR, Jensen VB, Javle M, Rodon J, Meric-Bernstam F. Co-clinical Trial of Novel Bispecific Anti-HER2 Antibody Zanidatamab in Patient-Derived Xenografts. Cancer Discov 2024; 14:828-845. [PMID: 38358339 PMCID: PMC11064988 DOI: 10.1158/2159-8290.cd-23-0838] [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: 08/18/2023] [Revised: 12/20/2023] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
Zanidatamab is a bispecific human epidermal growth factor receptor 2 (HER2)-targeted antibody that has demonstrated antitumor activity in a broad range of HER2-amplified/expressing solid tumors. We determined the antitumor activity of zanidatamab in patient-derived xenograft (PDX) models developed from pretreatment or postprogression biopsies on the first-in-human zanidatamab phase I study (NCT02892123). Of 36 tumors implanted, 19 PDX models were established (52.7% take rate) from 17 patients. Established PDXs represented a broad range of HER2-expressing cancers, and in vivo testing demonstrated an association between antitumor activity in PDXs and matched patients in 7 of 8 co-clinical models tested. We also identified amplification of MET as a potential mechanism of acquired resistance to zanidatamab and demonstrated that MET inhibitors have single-agent activity and can enhance zanidatamab activity in vitro and in vivo. These findings provide evidence that PDXs can be developed from pretreatment biopsies in clinical trials and may provide insight into mechanisms of resistance. SIGNIFICANCE We demonstrate that PDXs can be developed from pretreatment and postprogression biopsies in clinical trials and may represent a powerful preclinical tool. We identified amplification of MET as a potential mechanism of acquired resistance to the HER2 inhibitor zanidatamab and MET inhibitors alone and in combination as a therapeutic strategy. This article is featured in Selected Articles from This Issue, p. 695.
Collapse
Affiliation(s)
- Timothy P. DiPeri
- Department of Surgical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Kurt W. Evans
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Bailiang Wang
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Ming Zhao
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Argun Akcakanat
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, Texas
| | - Yasmeen Q. Rizvi
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, Texas
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, Texas
| | - Kaushik Varadarajan
- Department of Surgical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Burak Uzunparmak
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Ecaterina E. Dumbrava
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Paula R. Pohlmann
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
- Department of Breast Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - V. Behrana Jensen
- Department of Veterinary Medicine and Surgery, UT MD Anderson Cancer Center, Houston, Texas
| | - Milind Javle
- Department of Breast Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
43
|
Odintsov I, Makarem M, Nishino M, Bachert SE, Zhang T, LoPiccolo J, Paweletz CP, Gokhale PC, Ivanova E, Saldanha A, Rudin CM, Lockwood WW, Ladanyi M, Somwar R, Jänne PA, Sholl LM. Prevalence and Therapeutic Targeting of High-Level ERBB2 Amplification in NSCLC. J Thorac Oncol 2024; 19:732-748. [PMID: 38154514 PMCID: PMC11957432 DOI: 10.1016/j.jtho.2023.12.019] [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: 10/21/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
INTRODUCTION ERBB2 amplification in lung cancer remains poorly characterized. HER2 (encoded by ERBB2) is a transmembrane tyrosine kinase capable of ligand-independent dimerization and signaling when overexpressed, and a common cause of HER2 overexpression is ERBB2 amplification. Here, we evaluated the clinicopathologic and genomic characteristics of ERBB2-amplified NSCLC and explored a HER2 antibody-drug conjugate (ADC) therapeutic strategy. METHODS Our institutional next-generation DNA sequencing data (OncoPanel) from 5769 NSCLC samples (5075 patients) were queried for cases having high-level ERBB2 amplification (≥6 copies). Clinical and demographic characteristics were extracted from the electronic medical records. Efficacy of the pan-ERBB inhibitor afatinib or HER2 ADCs (trastuzumab deruxtecan and trastuzumab emtansine) was evaluated in NSCLC preclinical models and patients with ERBB2 amplification. RESULTS High-level ERBB2 amplification was identified in 0.9% of lung adenocarcinomas and reliably predicted overexpression of HER2. ERBB2 amplification events are detected in two distinct clinicopathologic and genomic subsets of NSCLC: as the sole mitogenic driver in tumors arising in patients with a smoking history or as a concomitant alteration with other mitogenic drivers in patients with a light or never smoking history. We further reveal that trastuzumab deruxtecan is effective therapy in in vitro and in vivo preclinical models of NSCLC harboring ERBB2 amplification and report two cases of clinical activity of an anti-HER2 ADC in patients who acquired ERBB2 amplification after previous targeted therapy. CONCLUSIONS High-level ERBB2 amplification reliably predicts HER2 overexpression in patients with NSCLC, and HER2 ADC is effective therapy in this population.
Collapse
Affiliation(s)
- Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maisam Makarem
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Emily Bachert
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Tom Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; School of Medicine, New York Medical College, Valhalla, New York
| | - Jaclyn LoPiccolo
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cloud P Paweletz
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Prafulla C Gokhale
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elena Ivanova
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Aisha Saldanha
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William W Lockwood
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
44
|
Fountzilas E, Tsimberidou AM, Hiep Vo H, Kurzrock R. Tumor-agnostic baskets to N-of-1 platform trials and real-world data: Transforming precision oncology clinical trial design. Cancer Treat Rev 2024; 125:102703. [PMID: 38484408 DOI: 10.1016/j.ctrv.2024.102703] [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: 01/08/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
Choosing the right drug(s) for the right patient via advanced genomic sequencing and multi-omic interrogation is the sine qua non of precision cancer medicine. Traditional cancer clinical trial designs follow well-defined protocols to evaluate the efficacy of new therapies in patient groups, usually identified by their histology/tissue of origin of their malignancy. In contrast, precision medicine seeks to optimize benefit in individual patients, i.e., to define who benefits rather than determine whether the overall group benefits. Since cancer is a disease driven by molecular alterations, innovative trial designs, including biomarker-defined tumor-agnostic basket trials, are driving ground-breaking regulatory approvals and deployment of gene- and immune-targeted drugs. Molecular interrogation further reveals the disruptive reality that advanced cancers are extraordinarily complex and individually distinct. Therefore, optimized treatment often requires drug combinations and N-of-1 customization, addressed by a new generation of N-of-1 trials. Real-world data and structured master registry trials are also providing massive datasets that are further fueling a transformation in oncology. Finally, machine learning is facilitating rapid discovery, and it is plausible that high-throughput computing, in silico modeling, and 3-dimensional printing may be exploitable in the near future to discover and design customized drugs in real time.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, St Luke's Clinic, Thessaloniki, Greece; European University Cyprus, German Oncology Center, Nicosia, Cyprus
| | - Apostolia-Maria Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA.
| | - Henry Hiep Vo
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, France; Medical College of Wisconsin, USA
| |
Collapse
|
45
|
Hattori M, Serelli-Lee V, Naito Y, Yamanaka T, Yasojima H, Nakamura R, Fujisawa T, Imai M, Nakamura Y, Bando H, Kawaguchi T, Yoshino T, Iwata H. Genomic Landscape of Circulating Tumor DNA in Patients With Hormone Receptor-Positive/Human Epidermal Growth Factor Receptor-2-Negative Metastatic Breast Cancer Treated With Abemaciclib: Data From the SCRUM-Japan Cancer Genome Screening Project. JCO Precis Oncol 2024; 8:e2300647. [PMID: 38635933 PMCID: PMC11161242 DOI: 10.1200/po.23.00647] [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: 11/21/2023] [Revised: 02/13/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024] Open
Abstract
PURPOSE To understand the mutational landscape of circulating tumor DNA (ctDNA) and tumor tissue of patients with hormone receptor-positive (HR+), human epidermal growth factor receptor-2-negative (HER2-) metastatic breast cancer (MBC) treated with abemaciclib + endocrine therapy (ET). METHODS Blood samples for ctDNA and/or tissue samples were collected from abemaciclib-treated patients with HR+/HER2- MBC enrolled in the SCRUM-Japan MONSTAR-SCREEN project. Blood samples were collected before abemaciclib initiation (baseline) and at disease progression/abemaciclib discontinuation (post abemaciclib treatment). Clinical and genomic characteristics including neoplastic burden (measured by shedding rate and maximum variant allele frequency [VAF]) were assessed at baseline. Genomic alterations in ctDNA were compared in paired baseline and post abemaciclib treatment samples. RESULTS All patients (N = 97) were female (median age, 57 years [IQR, 50-67]). In baseline ctDNA (n = 77), PIK3CA (37%), TP53 (28%), ESR1 (16%), and GATA3 (11%) were the most frequently mutated genes. Baseline tissue samples (n = 79) showed similar alteration frequencies. Among patients with baseline ctDNA data, 30% had received previous ET. ESR1 alteration frequency (35% v 8%; P < .01), median shedding rate (3 v 2), and maximum somatic VAF (4 v 0.8; both P < .05) were significantly higher in ctDNA from patients with previous ET than those without previous ET. In paired ctDNA samples (n = 33), PIK3CA and ESR1 alteration frequencies were higher after abemaciclib treatment than at baseline, though not statistically significant. Among the post-treatment alterations, those newly acquired were detected most frequently in FGF3/4/19 (18%); PIK3CA, TP53, CCND1, and RB1 (all 15%); and ESR1 (12%). CONCLUSION We summarized the ctDNA and cancer tissue mutational landscape, including overall neoplastic burden and PIK3CA and ESR1 hotspot mutations in abemaciclib-treated patients with HR+/HER2- MBC. The data provide insights that could help optimize treatment strategies in this population.
Collapse
Affiliation(s)
| | | | - Yoichi Naito
- National Cancer Center Hospital East, Chiba, Japan
| | | | - Hiroyuki Yasojima
- National Hospital Organization Osaka National Hospital, Osaka, Japan
| | | | | | - Mitsuho Imai
- National Cancer Center Hospital East, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Connolly RM, Wang V, Hyman DM, Grivas P, Mitchell EP, Wright JJ, Sharon E, Gray RJ, McShane LM, Rubinstein LV, Patton DR, Williams PM, Hamilton SR, Wang J, Wisinski KB, Tricoli JV, Conley BA, Harris LN, Arteaga CL, O'Dwyer PJ, Chen AP, Flaherty KT. Trastuzumab and Pertuzumab in Patients with Non-Breast/Gastroesophageal HER2-Amplified Tumors: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol J. Clin Cancer Res 2024; 30:1273-1280. [PMID: 38433347 PMCID: PMC10984755 DOI: 10.1158/1078-0432.ccr-23-0633] [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: 04/21/2023] [Revised: 07/05/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE NCI-MATCH assigned patients with advanced cancer and progression on prior treatment, based on genomic alterations in pretreatment tumor tissue. Arm J (EAY131-J) evaluated the combination of trastuzumab/pertuzumab (HP) across HER2-amplified tumors. PATIENTS AND METHODS Eligible patients had high levels of HER2 amplification [copy number (CN) ≥7] detected by central next-generation sequencing (NGS) or through NCI-designated laboratories. Patients with breast/gastroesophageal adenocarcinoma and those who received prior HER2-directed therapy were excluded. Enrollment of patients with colorectal cancer was capped at 4 based on emerging data. Patients received HP IV Q3 weeks until progression or unacceptable toxicity. Primary endpoint was objective response rate (ORR); secondary endpoints included progression-free survival (PFS) and overall survival (OS). RESULTS Thirty-five patients were enrolled, with 25 included in the primary efficacy analysis (CN ≥7 confirmed by a central lab, median CN = 28). Median age was 66 (range, 31-80), and half of all patients had ≥3 prior therapies (range, 1-11). The confirmed ORR was 12% [3/25 partial responses (colorectal, cholangiocarcinoma, urothelial cancers), 90% confidence interval (CI) 3.4%-28.2%]. There was one additional partial response (urothelial cancer) in a patient with an unconfirmed ERBB2 copy number. Median PFS was 3.3 months (90% CI 2.0-4.1), and median OS 9.4 months (90% CI 5.0-18.9). Treatment-emergent adverse events were consistent with prior studies. There was no association between HER2 CN and response. CONCLUSIONS HP was active in a selection of HER2-amplified tumors (non-breast/gastroesophageal) but did not meet the predefined efficacy benchmark. Additional strategies targeting HER2 and potential resistance pathways are warranted, especially in rare tumors.
Collapse
Affiliation(s)
- Roisin M Connolly
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, Maryland
- Cancer Research @UCC, College of Medicine and Health, University College Cork, Ireland
| | - Victoria Wang
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Petros Grivas
- University of Washington, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Edith P Mitchell
- Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - John J Wright
- Investigational Drug Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Elad Sharon
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Robert J Gray
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - Lisa M McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Larry V Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - David R Patton
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, Maryland
| | - P Mickey Williams
- Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | - Jue Wang
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, Texas
| | - Kari B Wisinski
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - James V Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Barbara A Conley
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Lyndsay N Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Carlos L Arteaga
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, Texas
| | | | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| |
Collapse
|
47
|
Kraus FBT, Sultova E, Heinrich K, Jung A, Westphalen CB, Tauber CV, Kumbrink J, Rudelius M, Klauschen F, Greif PA, König A, Chelariu-Raicu A, Czogalla B, Burges A, Mahner S, Wuerstlein R, Trillsch F. Genetics and beyond: Precision Medicine Real-World Data for Patients with Cervical, Vaginal or Vulvar Cancer in a Tertiary Cancer Center. Int J Mol Sci 2024; 25:2345. [PMID: 38397025 PMCID: PMC10888648 DOI: 10.3390/ijms25042345] [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: 01/14/2024] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Advances in molecular tumor diagnostics have transformed cancer care. However, it remains unclear whether precision oncology has the same impact and transformative nature across all malignancies. We conducted a retrospective analysis of patients with human papillomavirus (HPV)-related gynecologic malignancies who underwent comprehensive molecular profiling and subsequent discussion at the interdisciplinary Molecular Tumor Board (MTB) of the University Hospital, LMU Munich, between 11/2017 and 06/2022. We identified a total cohort of 31 patients diagnosed with cervical (CC), vaginal or vulvar cancer. Twenty-two patients (fraction: 0.71) harbored at least one mutation. Fifteen patients (0.48) had an actionable mutation and fourteen (0.45) received a recommendation for a targeted treatment within the MTB. One CC patient received a biomarker-guided treatment recommended by the MTB and achieved stable disease on the mTOR inhibitor temsirolimus for eight months. Factors leading to non-adherence to MTB recommendations in other patient cases included informed patient refusal, rapid deterioration, stable disease, or use of alternative targeted but biomarker-agnostic treatments such as antibody-drug conjugates or checkpoint inhibitors. Despite a remarkable rate of actionable mutations in HPV-related gynecologic malignancies at our institution, immediate implementation of biomarker-guided targeted treatment recommendations remained low, and access to targeted treatment options after MTB discussion remained a major challenge.
Collapse
Affiliation(s)
- Fabian B. T. Kraus
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Elena Sultova
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Andreas Jung
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - C. Benedikt Westphalen
- Department of Medicine III, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Christina V. Tauber
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Frederick Klauschen
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Philipp A. Greif
- Department of Medicine III, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany
| | - Alexander König
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Alexander Burges
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Rachel Wuerstlein
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| |
Collapse
|
48
|
Barzi A, Weipert CM, Espenschied CR, Raymond VM, Wang-Gillam A, Nezami MA, Gordon EJ, Mahadevan D, Mody K. ERBB2 (HER2) amplifications and co-occurring KRAS alterations in the circulating cell-free DNA of pancreatic ductal adenocarcinoma patients and response to HER2 inhibition. Front Oncol 2024; 14:1339302. [PMID: 38406801 PMCID: PMC10885695 DOI: 10.3389/fonc.2024.1339302] [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: 11/15/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Despite accumulating data regarding the genomic landscape of pancreatic ductal adenocarcinoma (PDAC), olaparib is the only biomarker-driven FDA-approved targeted therapy with a PDAC-specific approval. Treating ERBB2(HER2)-amplified PDAC with anti-HER2 therapy has been reported with mixed results. Most pancreatic adenocarcinomas have KRAS alterations, which have been shown to be a marker of resistance to HER2-targeted therapies in other malignancies, though the impact of these alterations in pancreatic cancer is unknown. We describe two cases of ERBB2-amplified pancreatic cancer patients treated with anti-HER2 therapy and provide data on the frequency of ERBB2 amplifications and KRAS alterations identified by clinical circulating cell-free DNA testing. Methods De-identified molecular test results for all patients with pancreatic cancer who received clinical cell-free circulating DNA analysis (Guardant360) between 06/2014 and 01/2018 were analyzed. Cell-free circulating DNA analysis included next-generation sequencing of up to 73 genes, including select small insertion/deletions, copy number amplifications, and fusions. Results Of 1,791 patients with pancreatic adenocarcinoma, 36 (2.0%) had an ERBB2 amplification, 26 (72.2%) of whom had a KRAS alteration. Treatment data were available for seven patients. Two were treated with anti-HER2 therapy after their cell-free circulating DNA result, with both benefiting from therapy, including one with a durable response to trastuzumab and no KRAS alteration detected until progression. Conclusion Our case series illustrates that certain patients with ERBB2-amplified pancreatic adenocarcinoma may respond to anti-HER2 therapy and gain several months of prolonged survival. Our data suggests KRAS mutations as a possible mechanism of primary and acquired resistance to anti-HER2 therapy in pancreatic cancer. Additional studies are needed to clarify the role of KRAS in resistance to anti-HER2 therapy.
Collapse
Affiliation(s)
- Afsaneh Barzi
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States
| | | | | | | | - Andrea Wang-Gillam
- Division of Oncology, Siteman Cancer Center, St. Louis, MO, United States
| | | | - Eva J. Gordon
- Private Health Management, Inc., Los Angeles, CA, United States
| | - Daruka Mahadevan
- Division of Hematology and Oncology, Department of Medicine, University of Texas Health, San Antonio, San Antonio, TX, United States
| | - Kabir Mody
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
49
|
Cienfuegos JA, Zozaya G, Rotellar F. Liver transplantation in colorectal metastases. Is there an indication for this procedure? REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:59-62. [PMID: 37539564 DOI: 10.17235/reed.2023.9873/2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Almost 25 % of patients with colorectal cancer present metastases at the time of diagnosis and 50 % go on to develop metastases in the course of the disease. Surgical resection is the only curative treatment although only between 20 % and 30 % of patients present resectable lesions. Although liver transplantation is contraindicated in unresectable metastases of colorectal cancer, ever since the publication of the results of a pilot study there has been renewed interest in transplantation in these patients. In two consecutive trials overall and recurrence-free 5-year survival rates of 83 % and 35 % respectively, have been reported, Currently several trials are ongoing which are expected to allow the patient selection criteria for the indication of liver transplantation to be refined.
Collapse
|
50
|
von Fritsch L, von Bubnoff N, Weber K, Kirfel J, Schreiber C, Keck T, Wellner U. Near complete remission of an inoperable pancreatic acinar cell carcinoma after BRAF-/MEK-inhibitor treatment-A case report and review of the literature. Genes Chromosomes Cancer 2024; 63:e23222. [PMID: 38340027 DOI: 10.1002/gcc.23222] [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: 09/04/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Pancreatic acinar cell carcinomas are rare malignant neoplasms. High-quality evidence about the best treatment strategy is lacking. We present the case of a 52-year-old male with a BRAFV600E -mutated PACC who experienced a complete remission after chemotherapy with BRAF-/MEK-inhibitors. CASE The patient presented with upper abdomen pain, night sweat, and weight loss. CT scan showed a pancreatic tumor extending from the pancreas head to body. Histological workup identified an acinar cell carcinoma. As the tumor was inoperable, chemotherapy with FOFIRNIOX was initiated and initially showed a slight regression of disease. The regimen had to be discontinued due to severe side effects. Molecular analysis identified a BRAFV600E mutation, so the patient was started on BRAF- and MEK-inhibitors (dabrafenib/trametinib). After 16 months, CT scans showed a near complete remission with a markedly improved overall health. DISCUSSION Studies suggest that up to one-fourth of PACCs carry a BRAF mutation and might therefore be susceptible to a BRAF-/MEK-inhibitor therapy. This offers a new therapeutic pathway to treat this rare but malignant neoplasm.
Collapse
Affiliation(s)
- Lennart von Fritsch
- Department of Surgery, University Hospital of Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
| | - Klaus Weber
- Luebecker Onkologische Schwerpunktpraxis, Lübeck, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
| | - Cleopatra Schreiber
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
| | - Tobias Keck
- Department of Surgery, University Hospital of Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
| | - Ulrich Wellner
- Department of Surgery, University Hospital of Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
| |
Collapse
|