1
|
Sztankovics D, Szalai F, Moldvai D, Dankó T, Scheich B, Pápay J, Sebestyén A, Krencz I. Comparison of molecular subtype composition between independent sets of primary and brain metastatic small cell lung carcinoma and matched samples. Lung Cancer 2025; 199:108071. [PMID: 39721126 DOI: 10.1016/j.lungcan.2024.108071] [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: 09/22/2024] [Revised: 11/14/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Recent advances in the subclassification of small cell lung carcinomas (SCLCs) may help to overcome the unmet need for targeted therapies and improve survival. However, limited information is available on how the expression of the subtype markers changes during tumour progression. Our study aimed to compare the expression of these markers in primary and brain metastatic SCLCs. MATERIALS AND METHODS Immunohistochemical analysis of the subtype markers was performed on 120 SCLCs (including 10 matched samples) and SCLC xenografts. RESULTS Compared to primary SCLCs, there was a significant increase in the proportion of mixed subtypes in brain metastases, with a rate of ASCL1high/NeuroD1high and ASCL1high/NeuroD1high/YAP1high subtypes increasing to 48 % and 18 %, respectively. The subtype of the paired samples matched in only one-third of the cases. Although we did not observe a significant change after chemotherapy, a continuous decrease in ASCL1 expression coupled with an increase in the NeuroD1 expression was detected in the xenografts in a long-term experiment. DISCUSSION Our results indicate that the expression of subtype markers frequently changes during disease progression, and subtype analysis of the primary SCLC may not provide accurate information about the characteristics of the recurrent or metastatic tumour. Therefore, repeated sampling and subtyping may be necessary for subtype-specific targeted therapy.
Collapse
Affiliation(s)
- Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Fatime Szalai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Titanilla Dankó
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Bálint Scheich
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary.
| |
Collapse
|
2
|
Ueki H, Jimbo N, Terakawa T, Hara T, Tobe T, Hirata J, Wakita N, Okamura Y, Suzuki K, Bando Y, Chiba K, Teishima J, Nakano Y, Miyake H. Evaluating RB1 and p53 as diagnostic markers in treatment-related neuroendocrine prostate cancer through immunohistochemistry and genomic analysis of RB1 and TP53. Prostate 2024; 84:1506-1514. [PMID: 39279246 DOI: 10.1002/pros.24791] [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: 07/01/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND The diagnosis of treatment-related neuroendocrine prostate cancer (t-NEPC) often involves a pathological assessment and immunohistochemistry (IHC) for neuroendocrine markers. Genomic alterations in RB1 and TP53 are frequently observed in NEPC and are believed to play a crucial role in the transformation of adenocarcinoma to NEPC. In this study, we examined the clinicopathologic, immunohistochemical, and genetic features of patients with t-NEPC to better understand their prognosis and diagnostic utility. METHODS This retrospective study reviewed the records of patients diagnosed with t-NEPC at Kobe University Hospital between October 2018 and December 2022. Clinical data, including age, serum neuroendocrine marker levels, and treatment history, were collected. IHC was performed for conventional neuroendocrine markers (synaptophysin, chromogranin A, and CD56) and RB1 and p53 expression. Next-generation sequencing (NGS) was conducted using FoundationOne® CDx to identify mutations in RB1 and TP53. RESULTS This study included 20 patients with t-NEPC. The median time from ADT initiation to development was 42.8 months. IHC revealed RB1 loss in 75% of cases and p53 abnormalities in 75% of cases. NGS identified RB1 mutations in 55% and TP53 mutations in 75% of cases. The concordance between NGS and IHC results was high, with 70% (14/20) agreement for RB1/RB1 and 80% (16/20) for p53/TP53. The immunostaining and genomic analysis of RB1/RB1 and p53/TP53 showed abnormal findings for the four negative cases for conventional neuroendocrine markers. CONCLUSIONS This study indicated high concordance between IHC and NGS findings for RB1/RB1 and p53/TP53 in t-NEPC. We provide a comprehensive benchmark of NGS performance compared with IHC, and these findings may help increase the diagnostic sensitivity of t-NEPC.
Collapse
Affiliation(s)
- Hideto Ueki
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoe Jimbo
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoaki Terakawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takuto Hara
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taisuke Tobe
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Junichiro Hirata
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoto Wakita
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyoshi Okamura
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kotaro Suzuki
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yukari Bando
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koji Chiba
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jun Teishima
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuzo Nakano
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideaki Miyake
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
3
|
Febres-Aldana CA, Elsayad MM, Saliba M, Bhanot U, Ntiamoah P, Takeyama A, Purgina BM, Rodriguez-Urrego PA, Marusic Z, Jakovcevic A, Chute DJ, Dunn LA, Ganly I, Cohen MA, Pfister DG, Ghossein RA, Baine MK, Rekhtman N, Dogan S. Analysis of ASCL1/NEUROD1/POU2F3/YAP1 Yields Novel Insights for the Diagnosis of Olfactory Neuroblastoma and Identifies Sinonasal Tuft Cell-Like Carcinoma. Mod Pathol 2024; 38:100674. [PMID: 39613231 DOI: 10.1016/j.modpat.2024.100674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/08/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
The diagnosis and treatment of sinonasal small round epithelial/neuroepithelial malignancies depend on the expression of conventional neuroendocrine markers (NEMs), such as synaptophysin, chromogranin A, INSM1, and CD56/NCAM1. However, these tumors remain diagnostically challenging because of overlapping histologic and immunohistochemical features. The transcriptional regulators ASCL1, NEUROD1, POU2F3, and YAP1 are novel NEM (nNEM) used for the subtyping of small-cell lung cancer (SCLC). Here, we assessed the immunoexpression of nNEM in 76 sinonasal malignancies, including 27 olfactory neuroblastomas (ONB), 14 small-cell neuroendocrine carcinomas (SCNEC), 2 large-cell neuroendocrine carcinomas, 12 sinonasal undifferentiated carcinomas (SNUC), 7 olfactory carcinomas (OC), 11 SWI/SNF-deficient carcinomas, and 3 neuroendocrine tumors. We correlated nNEM expression with the extent of neuroendocrine (NE) differentiation, as defined by averaged conventional NEM expression (NE-high: H-score, ≥150; NE-low: H-score, <150). Dominant NE subtypes were defined by the nNEM with the highest H-score. Coexpression of 2 nNEM with <100 H-score difference defined a codominant NE subtype. NE differentiation positively correlated with NEUROD1 and negatively with YAP1 expression (P < .0001). ONB were NE-high (96%), and all were NEUROD1-dominant/POU2F3-negative/ASCL1-negative (low)/YAP1-negative (low). In contrast to ONB, all OC were NE-low, mostly (71%) codominant subtypes, NEUROD1-low (negative) (100%, P = .0001), and YAP1 high (71%; P = .0001). Most notably, all SNUC were POU2F3-(co)dominant/NEUROD1-negative irrespective of the IDH2 mutations. Sinonasal tumors with high POU2F3 expression showed enrichment for "tuft cell carcinoma" and tuft cell signatures (P = .009). Similar to SCLC, SCNEC was heterogeneous in terms of nNEM expression comprising several molecular subtypes, including ASCL1-(co)dominant (43%) cases. All SWI/SNF-deficient carcinomas were consistently ASCL1/NEUROD1/POU2F3-negative and YAP1-positive. ASCL1/NEUROD1/POU2F3/YAP1 are useful markers in the differential diagnosis of ONB, SNUC, OC, and SWI/SNF-deficient carcinomas. Subsets of SNUC and large-cell neuroendocrine carcinomas may represent tuft cell-like carcinomas, suggesting that the tuft cell could be explored as the cell of origin for these tumors. The therapeutic vulnerabilities associated with POU2F3 expression in SCLC suggest that a similar approach might be considered for POU2F3-positive carcinomas of the sinonasal tract. Given their diagnostic and possible therapeutic relevance, nNEM have the potential to transform the way we approach the diagnosis and management of sinonasal small round epithelial/neuroepithelial malignancies.
Collapse
Affiliation(s)
- Christopher A Febres-Aldana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mahmoud M Elsayad
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maelle Saliba
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Umesh Bhanot
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter Ntiamoah
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anjanie Takeyama
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bibianna M Purgina
- Department of Pathology and Laboratory Medicine, Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Paula A Rodriguez-Urrego
- Department of Pathology, University Hospital of the Santa Fe de Bogotá Medical Foundation, Bogotá, Colombia
| | - Zlatko Marusic
- Clinical Department of Pathology and Cytology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Antonia Jakovcevic
- Clinical Department of Pathology and Cytology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Deborah J Chute
- Department of Pathology and Laboratory Medicine Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Lara A Dunn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc A Cohen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David G Pfister
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald A Ghossein
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marina K Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Snjezana Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
4
|
Vigdorovits A, Olteanu GE, Pascalau AV, Pirlog R, Berindan-Neagoe I, Pop OL. Novel Immunohistochemical Profiling of Small-Cell Lung Cancer: Correlations Between Tumor Subtypes and Immune Microenvironment. Diagnostics (Basel) 2024; 14:2660. [PMID: 39682568 DOI: 10.3390/diagnostics14232660] [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: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Small-cell lung cancer (SCLC) is a highly aggressive malignancy with an emerging molecular classification based on the expression of the transcription factors ASCL1, NEUROD1, and POU2F3. This study aimed to explore the relationship between these novel subtypes and the tumor immune microenvironment (TIME), particularly CD8+ and CD4+ tumor-infiltrating lymphocytes (TILs). METHODS In 51 cases of patients with SCLC, immunohistochemical (IHC) stains for ASCL1, NEUROD1, POU2F3, CD56, Ki67, CD8, and CD4 were performed. H-scores for the novel transcription factors were calculated to determine tumor subtype. CD8+ and CD4+ TIL counts were averaged across 10 high-power fields. The Kruskal-Wallis test and subsequent post hoc Dunn tests were used to determine the differences in transcription factor expression and TILs across subtypes. RESULTS In our cohort, 68.62% of our cases were SCLC-A, 9.80% were SCLC-N, 7.84% were SCLC-P, and 13.72% were SCLC-I. Significant differences were observed in the expression of ASCL1, NEUROD1, and POU2F3 across subtypes. CD8+ TILs were more abundant in SCLC-P and SCLC-I. CD8+ TILs were negatively correlated with ASCL1 expression (p < 0.05) and positively correlated with POU2F3 expression (p < 0.005). CONCLUSIONS This study highlights the need to integrate the novel SCLC classification with data regarding the TIME to better inform patient prognosis and treatment.
Collapse
Affiliation(s)
- Alon Vigdorovits
- Department of Morphological Disciplines, University of Oradea, 410087 Oradea, Romania
| | - Gheorghe-Emilian Olteanu
- British Columbia Cancer, Department of Pathology, Vancouver, BC V5Z 4E6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | | | - Radu Pirlog
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, 94010 Créteil, France
- INSERM U955, Université Paris Est Créteil, 94010 Créteil, France
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Doctoral School, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ovidiu-Laurean Pop
- Department of Morphological Disciplines, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
5
|
Ireland AS, Hawgood SB, Xie DA, Barbier MW, Lucas-Randolph S, Tyson DR, Zuo LY, Witt BL, Govindan R, Dowlati A, Moser JC, Puri S, Rudin CM, Chan JM, Elliott A, Oliver TG. Basal cell of origin resolves neuroendocrine-tuft lineage plasticity in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623500. [PMID: 39605338 PMCID: PMC11601426 DOI: 10.1101/2024.11.13.623500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Neuroendocrine and tuft cells are rare, chemosensory epithelial lineages defined by expression of ASCL1 and POU2F3 transcription factors, respectively1,2. Neuroendocrine cancers, including small cell lung cancer (SCLC), frequently display tuft-like subsets, a feature linked to poor patient outcomes3-13. The mechanisms driving neuroendocrine-tuft tumour heterogeneity, and the origins of tuft-like cancers are unknown. Using multiple genetically-engineered animal models of SCLC, we demonstrate that a basal cell of origin (but not the accepted neuroendocrine origin) generates neuroendocrine-tuft-like tumours that highly recapitulate human SCLC. Single-cell clonal analyses of basal-derived SCLC further uncovers unexpected transcriptional states and lineage trajectories underlying neuroendocrine-tuft plasticity. Uniquely in basal cells, introduction of genetic alterations enriched in human tuft-like SCLC, including high MYC, PTEN loss, and ASCL1 suppression, cooperate to promote tuft-like tumours. Transcriptomics of 944 human SCLCs reveal a basal-like subset and a tuft-ionocyte-like state that altogether demonstrate remarkable conservation between cancer states and normal basal cell injury response mechanisms14-18. Together, these data suggest that the basal cell is a plausible origin for SCLC and other neuroendocrine-tuft cancers that can explain neuroendocrine-tuft heterogeneity-offering new insights for targeting lineage plasticity.
Collapse
Affiliation(s)
- Abbie S. Ireland
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Sarah B. Hawgood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Daniel A. Xie
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Margaret W. Barbier
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | | | - Darren R. Tyson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Lisa Y. Zuo
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Benjamin L. Witt
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Ramaswamy Govindan
- Division of Oncology, Department of Medicine, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Afshin Dowlati
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | - Sonam Puri
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Charles M. Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Joseph M. Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Trudy G. Oliver
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| |
Collapse
|
6
|
Akbulut D, Whiting K, Teo MY, Tallman JE, Ozcan GG, Basar M, Jia L, Rammal R, Chen JF, Sarungbam J, Chen YB, Gopalan A, Fine SW, Tickoo SK, Mehra R, Baine M, Bochner BH, Pietzak EJ, Bajorin DF, Rosenberg JE, Iyer G, Solit DB, Reuter VE, Rekhtman N, Ostrovnaya I, Al-Ahmadie H. Differential NEUROD1, ASCL1, and POU2F3 Expression Defines Molecular Subsets of Bladder Small Cell/Neuroendocrine Carcinoma With Prognostic Implications. Mod Pathol 2024; 37:100557. [PMID: 38964503 PMCID: PMC11490389 DOI: 10.1016/j.modpat.2024.100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/22/2024] [Accepted: 06/18/2024] [Indexed: 07/06/2024]
Abstract
Small cell carcinomas (SMC) of the lung are now molecularly classified based on the expression of transcriptional regulators (NEUROD1, ASCL1, POU2F3, and YAP1) and DLL3, which has emerged as an investigational therapeutic target. PLCG2 has been shown to identify a distinct subpopulation of lung SMC with stem cell-like and prometastasis features and poor prognosis. We analyzed the expression of these novel neuroendocrine markers and their association with traditional neuroendocrine markers and patient outcomes in a cohort of bladder neuroendocrine carcinoma (NEC) consisting of 103 SMC and 19 large cell NEC (LCNEC) assembled in tissue microarrays. Coexpression patterns were assessed and integrated with detailed clinical annotation including overall (OS) and recurrence-free survival (RFS) and response to neoadjuvant/adjuvant chemotherapy. We identified 5 distinct molecular subtypes in bladder SMC based on the expression of ASCL1, NEUROD1, and POU2F3: ASCL1+/NEUROD1- (n = 33; 34%), ASCL1- /NEUROD1+ (n = 21; 21%), ASCL1+/NEUROD1+ (n = 17; 17%), POU2F3+ (n = 22, 22%), and ASCL1- /NEUROD1- /POU2F3- (n = 5, 5%). POU2F3+ tumors were mutually exclusive with those expressing ASCL1 and NEUROD1 and exhibited lower expression of traditional neuroendocrine markers. PLCG2 expression was noted in 33 tumors (32%) and was highly correlated with POU2F3 expression (P < .001). DLL3 expression was high in both SMC (n = 72, 82%) and LCNEC (n = 11, 85%). YAP1 expression was enriched in nonneuroendocrine components and negatively correlated with all neuroendocrine markers. In patients without metastatic disease who underwent radical cystectomy, PLCG2+ or POU2F3+ tumors had shorter RFS and OS (P < .05), but their expression was not associated with metastasis status or response to neoadjuvant/adjuvant chemotherapy. In conclusion, the NEC of the bladder can be divided into distinct molecular subtypes based on the expression of ASCL1, NEUROD1, and POU2F3. POU2F3-expressing tumors represent an ASCL1/NEUROD1-negative subset of bladder NEC characterized by lower expression of traditional neuroendocrine markers. Marker expression patterns were similar in SMC and LCNEC. Expression of PLCG2 and POU2F3 was associated with shorter RFS and OS. DLL3 was expressed at high levels in both SMC and LCNEC of the bladder, nominating it as a potential therapeutic target.
Collapse
MESH Headings
- Humans
- Basic Helix-Loop-Helix Transcription Factors/analysis
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Urinary Bladder Neoplasms/pathology
- Urinary Bladder Neoplasms/mortality
- Urinary Bladder Neoplasms/metabolism
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Neuroendocrine/therapy
- Male
- Female
- Aged
- Middle Aged
- Prognosis
- Carcinoma, Small Cell/pathology
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/mortality
- Carcinoma, Small Cell/genetics
- Tissue Array Analysis
- POU Domain Factors/genetics
- POU Domain Factors/metabolism
- POU Domain Factors/analysis
- Adult
- Aged, 80 and over
- Immunohistochemistry
- Disease-Free Survival
Collapse
Affiliation(s)
- Dilara Akbulut
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Karissa Whiting
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Min-Yuen Teo
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jacob E Tallman
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gamze Gokturk Ozcan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Merve Basar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Liwei Jia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, UT Southwestern, Dallas, Texas
| | - Rayan Rammal
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jie-Fu Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Judy Sarungbam
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ying-Bei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anuradha Gopalan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samson W Fine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Satish K Tickoo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Marina Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernard H Bochner
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eugene J Pietzak
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dean F Bajorin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan E Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gopa Iyer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David B Solit
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Victor E Reuter
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
7
|
Jiang J, Han D, Wang J, Wen W, Zhang R, Qin W. Neuroendocrine transdifferentiation in human cancer: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e761. [PMID: 39372390 PMCID: PMC11450264 DOI: 10.1002/mco2.761] [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: 07/17/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 10/08/2024] Open
Abstract
Neuroendocrine transdifferentiation (NEtD), also commonly referred to as lineage plasticity, emerges as an acquired resistance mechanism to molecular targeted therapies in multiple cancer types, predominately occurs in metastatic epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer treated with EGFR tyrosine kinase inhibitors and metastatic castration-resistant prostate cancer treated with androgen receptor targeting therapies. NEtD tumors are the lethal cancer histologic subtype with unfavorable prognosis and limited treatment. A comprehensive understanding of molecular mechanism underlying targeted-induced plasticity could greatly facilitate the development of novel therapies. In the past few years, increasingly elegant studies indicated that NEtD tumors share key the convergent genomic and phenotypic characteristics irrespective of their site of origin, but also embrace distinct change and function of molecular mechanisms. In this review, we provide a comprehensive overview of the current understanding of molecular mechanism in regulating the NEtD, including genetic alterations, DNA methylation, histone modifications, dysregulated noncoding RNA, lineage-specific transcription factors regulation, and other proteomic alterations. We also provide the current management of targeted therapies in clinical and preclinical practice.
Collapse
Affiliation(s)
- Jun Jiang
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
- Department of Health Service, Base of Health ServiceAir Force Medical UniversityXi'anChina
| | - Donghui Han
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| | - Jiawei Wang
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, and National Translational Science Center for Molecular MedicineAir Force Medical UniversityXi'anChina
| | - Weihong Wen
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical ResearchNorthwestern Polytechnical UniversityXi'anChina
| | - Rui Zhang
- State Key Laboratory of Cancer BiologyDepartment of ImmunologyAir Force Medical UniversityXi'anChina
| | - Weijun Qin
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| |
Collapse
|
8
|
Redin E, Quintanal-Villalonga Á, Rudin CM. Small cell lung cancer profiling: an updated synthesis of subtypes, vulnerabilities, and plasticity. Trends Cancer 2024; 10:935-946. [PMID: 39164163 DOI: 10.1016/j.trecan.2024.07.008] [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: 06/01/2024] [Revised: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Small cell lung cancer (SCLC) is a devastating disease with high proliferative and metastatic capacity. SCLC has been classified into molecular subtypes based on differential expression of lineage-defining transcription factors. Recent studies have proposed new subtypes that are based on both tumor-intrinsic and -extrinsic factors. SCLC demonstrates substantial intratumoral subtype heterogeneity characterized by highly plastic transcriptional states, indicating that the initially dominant subtype can shift during disease progression and in association with resistance to therapy. Strategies to promote or constrain plasticity and cell fate transitions have nominated novel targets that could prompt the development of more durably effective therapies for patients with SCLC. In this review, we describe the latest advances in SCLC subtype classification and their biological and clinical implications.
Collapse
Affiliation(s)
- Esther Redin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Álvaro Quintanal-Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology Program, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
9
|
Desai N, D'Ambrosio D, Dombrowski KS, Illei PB, Heymann JJ. Pulmonary Cytopathology: Current and Future Impact of Microscopy and Immunohistochemistry. Surg Pathol Clin 2024; 17:411-429. [PMID: 39129140 DOI: 10.1016/j.path.2024.06.003] [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: 08/13/2024]
Abstract
With the advancement of tissue procurement techniques, in-depth knowledge of morphology is crucial for cytopathologists to diagnose neoplastic and nonneoplastic lung diseases optimally. Cytopathologists must also be well versed in immunohistochemistry/immunocytochemistry markers and their interpretation for an accurate diagnosis.
Collapse
Affiliation(s)
- Niyati Desai
- Department of Pathology and Cell Biology, New York-Presbyterian Hospital-Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Danielle D'Ambrosio
- Department of Pathology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016, USA
| | - Katya S Dombrowski
- Department of Pathology, The Johns Hopkins University School of Medicine, 600 North Wolfe St., Baltimore, MD 21287, USA
| | - Peter B Illei
- Department of Pathology, The Johns Hopkins University School of Medicine, 600 North Wolfe St., Baltimore, MD 21287, USA
| | - Jonas J Heymann
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital-Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
10
|
Yan L, Zhao X, Chang L, Jiang H, Zhang Z. The Significance of Insulinoma-Associated Protein 1 in the Pathological Diagnosis of Small-Cell Lung Cancer in Biopsy Specimens. Int J Surg Pathol 2024; 32:1082-1088. [PMID: 38192158 DOI: 10.1177/10668969231215817] [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: 01/10/2024]
Abstract
Objective: Our purpose was to investigate the clinicopathological diagnostic value of immunohistochemical antibody for insulinoma-associated protein 1 (INSM1) in biopsy specimens of SCLC. Methods: Biopsy specimens of SCLC diagnosed at the pathology department of Tangshan Gongren Hospital from January 2022 to June 2023 were selected. INSM1 expression was detected and compared with conventional neuroendocrine markers synaptophysin (SYP), chromogranin A (CHGA), and CD56 regarding expression sensitivity and specificity. Results: The sensitivity of INSM1 expression was significantly higher than that of CHGA (95% vs 50%, P = .000), but there was no statistically significant difference in the specificity of INSM1, SYP, CHGA, and CD56 expression (100% vs 94% vs 98% vs 92%, respectively, P = .241, 1.000, .126). Conclusions: INSM1 antibody shows high sensitivity and specificity in the expression of SCLC and serves as a reliable immunohistochemical marker in the clinicopathological diagnosis of SCLC in biopsy specimens.
Collapse
Affiliation(s)
- Limin Yan
- Department of Pathology, Tangshan Gongren Hospital, Tangshan, Hebei, China
| | - Xueli Zhao
- Department of Pathology, Tangshan Gongren Hospital, Tangshan, Hebei, China
| | - Liming Chang
- Department of Pathology, Tangshan Gongren Hospital, Tangshan, Hebei, China
| | - Haixian Jiang
- Department of Pathology, Tangshan Gongren Hospital, Tangshan, Hebei, China
| | - Zhiyong Zhang
- Department of Pathology, Tangshan Gongren Hospital, Tangshan, Hebei, China
| |
Collapse
|
11
|
LIN Z, FAN L, HE P. [Molecular Subtype of Small Cell Lung Cancer:
Challenge for Transforming into Clinical Practice]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:605-612. [PMID: 39318253 PMCID: PMC11425680 DOI: 10.3779/j.issn.1009-3419.2024.106.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 09/26/2024]
Abstract
Small cell lung cancer (SCLC), one of the histological subtypes of lung cancer, is characterized by high proliferation, early metastasis, susceptibility to drug resistance and recurrence. For several years, SCLC has always been regarded as a homogeneous disease, treated with a unified radiotherapy and chemotherapy strategy. Despite significant early therapeutic effects, drug resistance and recurrence occur quickly, and there is a lack of satisfactory treatment results, which may be due to insufficient understanding of the tumor heterogeneity of SCLC at present. Recently, the concept of SCLC molecular subtype based on the definition of relatively high expression of lineage transcription factors has been proposed in preclinical studies. This article mainly elaborates on the current status and latest findings of SCLC molecular subtype, emphasizing the potential problems that molecular typing may encounter in clinical practice, aiming to promote understanding of the research progress of molecular subtype in SCLC.
.
Collapse
|
12
|
Duplaquet L, So K, Ying AW, Pal Choudhuri S, Li X, Xu GD, Li Y, Qiu X, Li R, Singh S, Wu XS, Hamilton S, Chien VD, Liu Q, Qi J, Somerville TDD, Heiling HM, Mazzola E, Lee Y, Zoller T, Vakoc CR, Doench JG, Forrester WC, Abrams T, Long HW, Niederst MJ, Drapkin BJ, Kadoch C, Oser MG. Mammalian SWI/SNF complex activity regulates POU2F3 and constitutes a targetable dependency in small cell lung cancer. Cancer Cell 2024; 42:1352-1369.e13. [PMID: 39029464 PMCID: PMC11494612 DOI: 10.1016/j.ccell.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/22/2024] [Accepted: 06/21/2024] [Indexed: 07/21/2024]
Abstract
Small cell lung cancers (SCLCs) are composed of heterogeneous subtypes marked by lineage-specific transcription factors, including ASCL1, NEUROD1, and POU2F3. POU2F3-positive SCLCs, ∼12% of all cases, are uniquely dependent on POU2F3 itself; as such, approaches to attenuate POU2F3 expression may represent new therapeutic opportunities. Here using genome-scale screens for regulators of POU2F3 expression and SCLC proliferation, we define mSWI/SNF complexes as top dependencies specific to POU2F3-positive SCLC. Notably, chemical disruption of mSWI/SNF ATPase activity attenuates proliferation of all POU2F3-positive SCLCs, while disruption of non-canonical BAF (ncBAF) via BRD9 degradation is effective in pure non-neuroendocrine POU2F3-SCLCs. mSWI/SNF targets to and maintains accessibility over gene loci central to POU2F3-mediated gene regulatory networks. Finally, clinical-grade pharmacologic disruption of SMARCA4/2 ATPases and BRD9 decreases POU2F3-SCLC tumor growth and increases survival in vivo. These results demonstrate mSWI/SNF-mediated governance of the POU2F3 oncogenic program and suggest mSWI/SNF inhibition as a therapeutic strategy for POU2F3-positive SCLCs.
Collapse
Affiliation(s)
- Leslie Duplaquet
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Kevin So
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA; Biological and Biomedical Sciences Graduate Program, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander W Ying
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shreoshi Pal Choudhuri
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xinyue Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Grace D Xu
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Yixiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Rong Li
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Shilpa Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Xiaoli S Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA
| | - Seth Hamilton
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Victor D Chien
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qi Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Hillary M Heiling
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Emanuele Mazzola
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Yenarae Lee
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thomas Zoller
- Novartis BioMedical Research, Cambridge, MA 02139, USA
| | | | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Tinya Abrams
- Novartis BioMedical Research, Cambridge, MA 02139, USA
| | - Henry W Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Benjamin J Drapkin
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cigall Kadoch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Matthew G Oser
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Peressini M, Garcia-Campelo R, Massuti B, Martí C, Cobo M, Gutiérrez V, Dómine M, Fuentes J, Majem M, de Castro J, Córdoba JF, Diz MP, Isla D, Esteban E, Carcereny E, Vila L, Moreno-Vega A, Ros S, Moreno A, García FJ, Huidobro G, Aguado C, Cebey-López V, Valdivia J, Palmero R, Lianes P, López-Brea M, Vidal OJ, Provencio M, Arriola E, Baena J, Herrera M, Bote H, Molero M, Adradas V, Ponce-Aix S, Nuñez-Buiza A, Ucero Á, Hernandez S, Lopez-Rios F, Conde E, Paz-Ares L, Zugazagoitia J. Spatially Preserved Multi-Region Transcriptomic Subtyping and Biomarkers of Chemoimmunotherapy Outcome in Extensive-Stage Small Cell Lung Cancer. Clin Cancer Res 2024; 30:3036-3049. [PMID: 38630755 DOI: 10.1158/1078-0432.ccr-24-0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/12/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Transcriptomic subtyping holds promise for personalized therapy in extensive-stage small cell lung cancer (ES-SCLC). In this study, we aimed to assess intratumoral transcriptomic subtype diversity and to identify biomarkers of long-term chemoimmunotherapy benefit in human ES-SCLC. EXPERIMENTAL DESIGN We analyzed tumor samples from 58 patients with ES-SCLC enrolled in two multicenter single-arm phase IIIb studies evaluating frontline chemoimmunotherapy in Spain: n = 32 from the IMfirst trial and n = 26 from the CANTABRICO trial. We used the GeoMx Digital Spatial Profiler system to perform multi-region transcriptomic analysis. For subtype classification, we performed hierarchical clustering using the relative expression of ASCL1 (SCLC-A), NEUROD1 (SCLC-N), POU2F3 (SCLC-P), and YAP1 (SCLC-Y). RESULTS Subtype distribution was found to be similar between bothcohorts, except for SCLC-P, which was not identified in the CANTABRICO_DSP cohort. A total of 44% of the patients in both cohorts had tumors with multiple coexisting transcriptional subtypes. Transcriptional subtypes or subtype heterogeneity was not associated with outcomes. Most potential targets did not show subtype-specific expression. Consistently in both cohorts, tumors from patients with long-term benefit (time to progression ≥12 months) contained an IFNγ-dominated mRNA profile, including enhanced capacity for antigen presentation. Hypoxia and glycolytic pathways were associated with resistance to chemoimmunotherapy. CONCLUSIONS This work suggests that intratumoral heterogeneity, inconsistent association with outcome, and unclear subtype-specific target expression might be significant challenges for subtype-based precision oncology in SCLC. Preexisting IFNγ-driven immunity and mitochondrial metabolism seem to be correlates of long-term efficacy in this study, although the absence of a chemotherapy control arm precludes concluding that these are predictive features specific for immunotherapy.
Collapse
Affiliation(s)
- Melina Peressini
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | | | - Bartomeu Massuti
- Department of Medical Oncology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Cristina Martí
- Department of Medical Oncology, Hospital Universitario Sant Joan de Reus, Tarragona, Spain
| | - Manuel Cobo
- Department of Medical Oncology, UGC intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Vanesa Gutiérrez
- Department of Medical Oncology, Complejo Hospitalario Regional de Málaga, Málaga, Spain
| | - Manuel Dómine
- Department of Medical Oncology, Hospital Universitario Fundación Jimenez Díaz, Madrid, Spain
| | - José Fuentes
- Department of Medical Oncology, Hospital Universitario Virgen de Valme, Sevilla, Spain
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Javier de Castro
- Department of Medical Oncology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Juan F Córdoba
- Department of Medical Oncology, Hospital Universitario Arnau de Villanova, Lérida, Spain
| | - María P Diz
- Department of Medical Oncology, Complejo Asistencial Universitario de León, León, Spain
| | - Dolores Isla
- Department of Medical Oncology, Hospital Universitario Lozano Blesa, Zaragoza, Spain
| | - Emilio Esteban
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Asturias, Spain
| | - Enric Carcereny
- Department of Medical Oncology, Instituto Catalán de Oncología de Badalona, Badalona-Applied Research Group in Oncology (B-ARGO), Barcelona, Spain
| | - Laia Vila
- Department of Medical Oncology, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Moreno-Vega
- Department of Medical Oncology, Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Silverio Ros
- Department of Medical Oncology, Hospital Universitario Virgen de La Arrixaca, Murcia, Spain
| | - Amaia Moreno
- Department of Medical Oncology, Hospital de Galdakao, Vizcaya, Spain
| | - Francisco J García
- Department of Medical Oncology, Hospital Universitario Son Llatzer, Mallorca, Spain
| | - Gerardo Huidobro
- Department of Medical Oncology, Hospital Universitario Álvaro Cunqueiro, Vigo, Spain
| | - Carlos Aguado
- Department of Medical Oncology, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Victor Cebey-López
- Department of Medical Oncology, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Javier Valdivia
- Department of Medical Oncology, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Ramón Palmero
- Department of Medical Oncology, Instituto Catalán de Oncología de Hospitalet de Llobregat, Barcelona, Spain
| | - Pilar Lianes
- Department of Medical Oncology, Hospital de Mataró, Barcelona, Spain
| | - Marta López-Brea
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Oscar J Vidal
- Department of Medical Oncology, Hospital Universitario La Fe, Valencia, Spain
| | - Mariano Provencio
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Edurne Arriola
- Department of Medical Oncology, Hospital del Mar, Barcelona, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Baena
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Mercedes Herrera
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Helena Bote
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Magdalena Molero
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Vera Adradas
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Santiago Ponce-Aix
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Angel Nuñez-Buiza
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Álvaro Ucero
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Department of Physiology, Complutense University, Madrid, Spain
| | - Susana Hernandez
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Lopez-Rios
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Complutense University, Madrid, Spain
| | - Esther Conde
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Complutense University, Madrid, Spain
| | - Luis Paz-Ares
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Department of Physiology, Complutense University, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| | - Jon Zugazagoitia
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
14
|
Ceresoli GL, Rossi G, Agustoni F, Bonomi L, Borghetti P, Bulotta A, Casartelli C, Cerea G, Colonese F, Del Signore E, Finocchiaro G, Gianoncelli L, Grisanti S, Maiolani M, Pagni F, Proto C, Rijavec E, Vittimberga I, Arcangeli S, Filippi AR. Management of patients with extensive small-cell lung cancer in the immunotherapy era: An Italian consensus through a Delphi approach. Crit Rev Oncol Hematol 2024; 199:104247. [PMID: 38307393 DOI: 10.1016/j.critrevonc.2023.104247] [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: 01/30/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Immunotherapy represented a turning point for treating extensive small-cell lung cancer (ES-SCLC). Although, many issues remain debated. METHODS A group of Italian medical and radiation oncologists with expertise in managing patients with ES-SCLC developed a list of statements divided in six areas of interest. The Delphi method was used to assess the consensus on the defined list of statements. RESULTS 32 statements were included in the final list to be voted by the Delphi panel, and 26 reached a consensus on the agreement. A prompt involvement of a multidisciplinary team is a priority to provide an integrated treatment strategy. First-line recommended treatment is immunotherapy in combination with platinum-based chemotherapy and etoposide for four cycles followed by maintenance immunotherapy. CONCLUSIONS While awaiting new data from clinical trials and real-world studies, these recommendations can represent a useful tool to guide the management of ES-SCLC patients in daily practice.
Collapse
Affiliation(s)
| | - Giulio Rossi
- Pathology Unit, Hospital Institute Fondazione Poliambulanza, Via Bissolati 57, 25124 Brescia, Italy
| | - Francesco Agustoni
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy; Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lucia Bonomi
- Unit of Oncology, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Paolo Borghetti
- Radiation Oncology Department, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Alessandra Bulotta
- Department of Oncology, IRCCS San Raffaele, via Olgettina 60, Milan, Italy
| | | | - Giulio Cerea
- Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | | | - Ester Del Signore
- Division of Thoracic Oncology, European Institute of Oncology, IEO, Milan, Italy
| | - Giovanna Finocchiaro
- Medical Oncology and Hematologic Unit, Humanitas Cancer Center, Istituto Clinico Humanitas-IRCCS, Rozzano, Italy
| | - Letizia Gianoncelli
- Medical Oncology Unit, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Salvatore Grisanti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Martina Maiolani
- U.O.C Oncologia Medica ASST Valtellina e Alto Lario, Sondrio, Italy
| | - Fabio Pagni
- Pathology, Department of Medicine and Surgery, University Milan Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Claudia Proto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Erika Rijavec
- Unit of Medical Oncology, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | | | - Stefano Arcangeli
- Department of Radiation Oncology, University of Milan Bicocca, Milan, Italy
| | - Andrea Riccardo Filippi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
15
|
Finlay JB, Ireland AS, Hawgood SB, Reyes T, Ko T, Olsen RR, Abi Hachem R, Jang DW, Bell D, Chan JM, Goldstein BJ, Oliver TG. Olfactory neuroblastoma mimics molecular heterogeneity and lineage trajectories of small-cell lung cancer. Cancer Cell 2024; 42:1086-1105.e13. [PMID: 38788720 PMCID: PMC11186085 DOI: 10.1016/j.ccell.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/13/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024]
Abstract
The olfactory epithelium undergoes neuronal regeneration from basal stem cells and is susceptible to olfactory neuroblastoma (ONB), a rare tumor of unclear origins. Employing alterations in Rb1/Trp53/Myc (RPM), we establish a genetically engineered mouse model of high-grade metastatic ONB exhibiting a NEUROD1+ immature neuronal phenotype. We demonstrate that globose basal cells (GBCs) are a permissive cell of origin for ONB and that ONBs exhibit cell fate heterogeneity that mimics normal GBC developmental trajectories. ASCL1 loss in RPM ONB leads to emergence of non-neuronal histopathologies, including a POU2F3+ microvillar-like state. Similar to small-cell lung cancer (SCLC), mouse and human ONBs exhibit mutually exclusive NEUROD1 and POU2F3-like states, an immune-cold tumor microenvironment, intratumoral cell fate heterogeneity comprising neuronal and non-neuronal lineages, and cell fate plasticity-evidenced by barcode-based lineage tracing and single-cell transcriptomics. Collectively, our findings highlight conserved similarities between ONB and neuroendocrine tumors with significant implications for ONB classification and treatment.
Collapse
Affiliation(s)
- John B Finlay
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham 27710, NC, USA
| | - Abbie S Ireland
- Department of Pharmacology and Cancer Biology, Duke University, Durham 27710, NC, USA
| | - Sarah B Hawgood
- Department of Pharmacology and Cancer Biology, Duke University, Durham 27710, NC, USA
| | - Tony Reyes
- Department of Pharmacology and Cancer Biology, Duke University, Durham 27710, NC, USA; Department of Oncological Sciences, University of Utah, Salt Lake City 84112, UT, USA
| | - Tiffany Ko
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham 27710, NC, USA
| | - Rachelle R Olsen
- Department of Oncological Sciences, University of Utah, Salt Lake City 84112, UT, USA
| | - Ralph Abi Hachem
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham 27710, NC, USA
| | - David W Jang
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham 27710, NC, USA
| | - Diana Bell
- Division of Anatomic Pathology, City of Hope Comprehensive Cancer Center, Duarte 91010, CA, USA
| | - Joseph M Chan
- Human Oncology and Pathogenesis Program, Memorial-Sloan Kettering Cancer Center, New York City 10065, NY, USA
| | - Bradley J Goldstein
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham 27710, NC, USA; Department of Neurobiology, Duke University, Durham 27710, NC, USA.
| | - Trudy G Oliver
- Department of Pharmacology and Cancer Biology, Duke University, Durham 27710, NC, USA; Department of Oncological Sciences, University of Utah, Salt Lake City 84112, UT, USA.
| |
Collapse
|
16
|
Cai Z, Cheng X, Liao S, Zou W, Li L, Liu F, Huang W. POU2F3-positive small cell carcinoma of the bladder: A clinicopathologic analysis of 4 cases and literature review. Pathol Res Pract 2024; 257:155296. [PMID: 38615507 DOI: 10.1016/j.prp.2024.155296] [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: 12/16/2023] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
POU class 2 homeobox 3 (POU2F3)-positive small cell bladder carcinoma (SCBC) is an extremely rare entity, and its clinicopathologic features have not been fully described. Here, we investigated the clinicopathologic features of 4 cases of POU2F3-positive small cell bladder carcinoma (SCBC) and reviewed the literature. We collected 12 cases of SCBC from our departmental archives and detected the expression of POU2F3 by immunohistochemical (IHC) staining. Selected cases with or without POU2F3 expression were subjected to gene expression analysis between two different groups using DESeq2 software. We identified 4 POU2F3-positive SCBC patients, 2 males and 2 females, with a mean age of 77 years. Three patients had hematuria, and 1 patient had dysuria. Radiologic findings showed a bladder mass. Pathologic diagnosis showed that 3 cases were pure SCBC and 1 was mixed urothelial cancer (UC). Histopathologically, four POU2F3-positive SCBC tumors were composed of small round cells with sparse cytoplasm, the nuclei were salt-and-pepper-like or finely granular. Tumor cells showed characteristic cytoplasmic staining with punctate positive signals for cytokeratin. Syn and CD56 were diffusely positive in all the 4 patients. CgA was positive in only one patient. POU2F3-positive SCBC showed higher expression levels of POU2F3, HMGA2 and PLCG2 genes by RNA-Seq. Our data showed the specific clinicopathologic features of 4 rare POU2F3-positive SCBC cases, and the distinct molecular feature was observed between POU2F3-positive and negative SCBC in the limited number of cases.
Collapse
Affiliation(s)
- Zhe Cai
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiuzhi Cheng
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shousheng Liao
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wanwan Zou
- Department of Pathology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Lixiang Li
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fanrong Liu
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wenyong Huang
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
17
|
Akbulut D, Al-Ahmadie H. Updates on Urinary Bladder Tumors With Neuroendocrine Features. Adv Anat Pathol 2024; 31:169-177. [PMID: 38523484 PMCID: PMC11006587 DOI: 10.1097/pap.0000000000000433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
The most common neuroendocrine tumor in the urinary bladder is small cell carcinoma, which can be pure or mixed with components of urothelial or other histologic subtypes. Large cell neuroendocrine carcinoma of the bladder is rare and remains ill-defined but is increasingly recognized. Well-differentiated neuroendocrine tumor and paraganglioma can arise in the bladder but are very rare in this location. Recent advances in molecular characterization allowed for better classification and may offer improved stratification of these tumors.
Collapse
Affiliation(s)
- Dilara Akbulut
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, MD
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center New York, NY
| |
Collapse
|
18
|
Cai Z, Zuo L, Hu F, You H, Lu X, Liao S, Liu F, Li L, Huang W. Abnormal expression of LCA and CD43 in SCLC: a rare case report and brief literature review. BMC Pulm Med 2024; 24:195. [PMID: 38649942 PMCID: PMC11036674 DOI: 10.1186/s12890-024-03005-w] [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: 05/28/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND To present an unusual case of abnormal LCA expression and CD43 in SCLC and to review the reported literature to avoid potential diagnostic pitfalls. CASE PRESENTATION A 73-year-old male patient suffered from persistent back pain for more than one month. MRI revealed a compression fracture of the L1-L5 vertebra. A CT scan revealed multiple nodules and masses at the left root of the neck, lung hilum and mediastinum, and multiple areas of bony destruction of the ribs. Histology of the tumor revealed that small and round cells were arranged in nests with areas of necrosis. The tumor cells were round to ovoid with scant cytoplasm and indistinct cell borders. The nuclear chromatin was finely granular, and the nucleoli were absent or inconspicuous. Immunohistochemically, the tumor cells were positive for cytokeratin, TTF-1, POU2F3, LCA, and CD43. CONCLUSION This report highlights a potential diagnostic pitfall in the diagnosis of SCLC, urges pathologists to exercise caution in cases of LCA and CD43 positivity and illustrates the need for further immunohistochemical studies to avoid misdiagnosis.
Collapse
Affiliation(s)
- Zhe Cai
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Linwei Zuo
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Fangfang Hu
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Huiyan You
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Xiangtong Lu
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Shousheng Liao
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Fanrong Liu
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Lixiang Li
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China
| | - Wenyong Huang
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, 330000, Nanchang, China.
| |
Collapse
|
19
|
Hiatt JB, Doebley AL, Arnold HU, Adil M, Sandborg H, Persse TW, Ko M, Wu F, Quintanal Villalonga A, Santana-Davila R, Eaton K, Dive C, Rudin CM, Thomas A, Houghton AM, Ha G, MacPherson D. Molecular phenotyping of small cell lung cancer using targeted cfDNA profiling of transcriptional regulatory regions. SCIENCE ADVANCES 2024; 10:eadk2082. [PMID: 38598634 PMCID: PMC11006233 DOI: 10.1126/sciadv.adk2082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 02/27/2024] [Indexed: 04/12/2024]
Abstract
We report an approach for cancer phenotyping based on targeted sequencing of cell-free DNA (cfDNA) for small cell lung cancer (SCLC). In SCLC, differential activation of transcription factors (TFs), such as ASCL1, NEUROD1, POU2F3, and REST defines molecular subtypes. We designed a targeted capture panel that identifies chromatin organization signatures at 1535 TF binding sites and 13,240 gene transcription start sites and detects exonic mutations in 842 genes. Sequencing of cfDNA from SCLC patient-derived xenograft models captured TF activity and gene expression and revealed individual highly informative loci. Prediction models of ASCL1 and NEUROD1 activity using informative loci achieved areas under the receiver operating characteristic curve (AUCs) from 0.84 to 0.88 in patients with SCLC. As non-SCLC (NSCLC) often transforms to SCLC following targeted therapy, we applied our framework to distinguish NSCLC from SCLC and achieved an AUC of 0.99. Our approach shows promising utility for SCLC subtyping and transformation monitoring, with potential applicability to diverse tumor types.
Collapse
Affiliation(s)
- Joseph B. Hiatt
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Veterans Affairs Puget Sound Healthcare System - Seattle Branch, Seattle, WA, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Anna-Lisa Doebley
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Henry U. Arnold
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mohamed Adil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Holly Sandborg
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas W. Persse
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Minjeong Ko
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alvaro Quintanal Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rafael Santana-Davila
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Keith Eaton
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Caroline Dive
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Charles M. Rudin
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Graduate Program in Pharmacology, Weill Cornell Medical College; New York, NY, USA
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - A. McGarry Houghton
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Gavin Ha
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
20
|
Kim G, Kim M, Nam EJ, Lee JY, Park E. Application of Small Cell Lung Cancer Molecular Subtyping Markers to Small Cell Neuroendocrine Carcinoma of the Cervix: NEUROD1 as a Poor Prognostic Factor. Am J Surg Pathol 2024; 48:364-372. [PMID: 37981832 DOI: 10.1097/pas.0000000000002155] [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: 11/21/2023]
Abstract
Cervical small cell neuroendocrine carcinoma (CSCNEC) is a rare, aggressive type of cervical cancer. The treatment for CSCNEC follows the chemotherapeutic regimens used for small cell lung cancer (SCLC), with which it shares similar clinical and histologic features. For the first time, we applied neuroendocrine (NE) and SCLC molecular subtyping immunohistochemical markers [achaete-scute homolog 1 (ASCL1), neurogenic differentiation factor 1 (NEUROD1), POU class 2 homeobox 3 (POU2F3), and yes-associated protein 1] in 45 patients with CSCNEC. For the combined NE score, 51.1% of NE-high and 48.9% of NE-low subtypes were identified. The NE-high subtype tended to show worse progression-free survival and overall survival (OS) than the NE-low subtype ( P =0.059 and P =0.07, respectively). Applying the SCLC molecular subtyping, 53.3% of cases were identified as NEUROD1-dominant, 17.8% as ASCL1-dominant, 13.3% as YAP-dominant, and 4.4% as POU2F3-dominant, while 11.1% of cases showed negative expression for all markers; the distribution was different from that of SCLC. The NEUROD1-dominant subtype exhibited the worst OS, while the POU2F3 subtype exhibited the best OS ( P =0.003), similar to SCLC. In addition, the ASCL1-dominant and NEUROD1-dominant subtypes showed high NE scores, while yes-associated protein 1-dominant and POU2F3-dominant subtypes showed low NE scores ( P =0.008). In multivariate analysis, the NEUROD1 expression was further identified as the independent prognostic factor for worse OS, together with the high FIGO stage. CSCNEC was revealed to be a heterogeneous disease with different biological phenotypes and to share some similarities and differences with SCLC. Regarding the ongoing development of tailored treatments based on biomarkers in SCLC, the application of biomarker-driven individualized therapy would improve clinical outcomes in patients with CSCNEC.
Collapse
Affiliation(s)
- Gilhyang Kim
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Milim Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Ji Nam
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunhyang Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Iida Y, Wakuda K, Kawata T, Morita M, Sekikawa M, Doshita K, Yabe M, Kodama H, Miura K, Morikawa N, Mamesaya N, Kobayashi H, Ko R, Ono A, Kenmotsu H, Naito T, Murakami H, Gon Y, Takahashi T. Relationship between patterns of immunohistochemical conventional neuroendocrine markers and efficacy of immune check point inhibitors in patients with extensive disease small cell lung cancer. Thorac Cancer 2024; 15:477-485. [PMID: 38243641 PMCID: PMC10883854 DOI: 10.1111/1759-7714.15218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Which patients benefit from the addition of immune checkpoint inhibitors (ICIs) to chemotherapy for small cell lung cancer (SCLC) remains unclear. There have been few reports on the efficacy of ICIs based on conventional immunohistochemical neuroendocrine (NE) markers (synaptophysin, chromogranin A, and neural cell adhesion molecule [NCAM]). In the present study, we aimed to analyze the relationship between the expression of immunohistochemical NE markers and the efficacy of ICIs in patients with extensive disease (ED)-SCLC, to assess whether conventional NE markers are predictive of ICIs. METHODS Patients with untreated ED-SCLC who received first-line therapy at the Shizuoka Cancer Center between November 2002 and July 2021 were retrospectively reviewed. We evaluated the efficacy of first-line chemotherapy according to the expression status of each immunohistochemical NE marker in patients treated with ICI plus chemotherapy (ICI-chemo group) and with chemotherapy alone (chemo group). RESULTS A total of 227 patients were included in the ICI-chemo and chemo groups, respectively. The progression-free survival (PFS) tended to be better in patients in the ICI-chemo group than those treated with chemotherapy alone in patients with NE marker-positive SCLC. In particular, it was statistically significant in patients with chromogranin A-positive SCLC (p = 0.036). In patients with NE marker-negative SCLC, no significant differences were observed in PFS between the two groups. There were no significant differences in overall survival (OS), regardless of the expression of any conventional NE marker. CONCLUSION Our study suggests that the efficacy of ICIs in addition to chemotherapy may be poor in patients with NE marker-negative SCLC.
Collapse
Affiliation(s)
- Yuko Iida
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
- Division of Respiratory Medicine, Department of Internal MedicineNihon University School of MedicineTokyoJapan
| | - Kazushige Wakuda
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Takuya Kawata
- Division of PathologyShizuoka Cancer CenterShizuokaJapan
| | - Meiko Morita
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Motoki Sekikawa
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Kosei Doshita
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Michitoshi Yabe
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Hiroaki Kodama
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Keita Miura
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Noboru Morikawa
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Nobuaki Mamesaya
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Haruki Kobayashi
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Ryo Ko
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Akira Ono
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | | | - Tateaki Naito
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | | | - Yasuhiro Gon
- Division of Respiratory Medicine, Department of Internal MedicineNihon University School of MedicineTokyoJapan
| | | |
Collapse
|
22
|
Chiang CL, Huang HC, Luo YH, Shen CI, Chao HS, Tseng YH, Chou TY, Schrump DS, Yeh YC, Chen YM. Clinical utility of immunohistochemical subtyping in patients with small cell lung cancer. Lung Cancer 2024; 188:107473. [PMID: 38232600 DOI: 10.1016/j.lungcan.2024.107473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/17/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
OBJECTIVES Molecular subtyping of small cell lung cancer (SCLC) tumors based on the expression of four transcription factors (ASCL1, NEUROD1, POU2F3, and YAP1) using immunohistochemical (IHC) staining has recently emerged as a proposed approach. This study was aimed to examine this subtyping method in Asian patients with SCLC and investigate its correlation with treatment efficacy. MATERIALS AND METHODS Seventy-two tumor samples from patients with SCLC, including de novo cases and those transformed from EGFR-mutant tumors, were analyzed. IHC staining was used to measure the expression of the four transcription factors and conventional SCLC markers. Subtypes were defined based on relative expression levels. The treatment response and outcome of patients receiving immune checkpoint inhibitors and chemotherapy were also reviewed. RESULTS ASCL1 was the most common subtype, observed in 55.2 % of the samples, followed by NEUROD1 (26.9 %) and POU2F3 (9 %). No tumor exhibited predominant YAP1 positivity, while 41.8 % of the samples demonstrated positivity for two subtype markers. Approximately 50 % of the patients experienced a subtype switch after disease progression. Patients with the ASCL1/NEUROD1 (SCLC-A/N) subtype had similar progression-free survival (PFS) compared to non-SCLC-A/N patients after treatment with immune checkpoint inhibitors plus chemotherapy. Transformed SCLC patients had significantly worse PFS than de novo SCLC patients after chemoimmunotherapy. (2.1 vs. 5.4 months, P = 0.023) CONCLUSIONS: This study revealed the challenges associated with using IHC alone for molecular subtyping, highlighting the frequent co-expression of subtypes and temporal changes following treatment. Further research is warranted to explore the prognostic and therapeutic implications of IHC subtyping in patients with SCLC.
Collapse
Affiliation(s)
- Chi-Lu Chiang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsu-Ching Huang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yung-Hung Luo
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-I Shen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Heng-Sheng Chao
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Teh-Ying Chou
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - David S Schrump
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yi-Chen Yeh
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
23
|
Oser MG, MacPherson D, Oliver TG, Sage J, Park KS. Genetically-engineered mouse models of small cell lung cancer: the next generation. Oncogene 2024; 43:457-469. [PMID: 38191672 PMCID: PMC11180418 DOI: 10.1038/s41388-023-02929-7] [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: 10/19/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/10/2024]
Abstract
Small cell lung cancer (SCLC) remains the most fatal form of lung cancer, with patients in dire need of new and effective therapeutic approaches. Modeling SCLC in an immunocompetent host is essential for understanding SCLC pathogenesis and ultimately discovering and testing new experimental therapeutic strategies. Human SCLC is characterized by near universal genetic loss of the RB1 and TP53 tumor suppressor genes. Twenty years ago, the first genetically-engineered mouse model (GEMM) of SCLC was generated using conditional deletion of both Rb1 and Trp53 in the lungs of adult mice. Since then, several other GEMMs of SCLC have been developed coupling genomic alterations found in human SCLC with Rb1 and Trp53 deletion. Here we summarize how GEMMs of SCLC have contributed significantly to our understanding of the disease in the past two decades. We also review recent advances in modeling SCLC in mice that allow investigators to bypass limitations of the previous generation of GEMMs while studying new genes of interest in SCLC. In particular, CRISPR/Cas9-mediated somatic gene editing can accelerate how new genes of interest are functionally interrogated in SCLC tumorigenesis. Notably, the development of allograft models and precancerous precursor models from SCLC GEMMs provides complementary approaches to GEMMs to study tumor cell-immune microenvironment interactions and test new therapeutic strategies to enhance response to immunotherapy. Ultimately, the new generation of SCLC models can accelerate research and help develop new therapeutic strategies for SCLC.
Collapse
Affiliation(s)
- Matthew G Oser
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - David MacPherson
- Division of Human Biology, Fred Hutch Cancer Center, Seattle, WA, 98109, USA
| | - Trudy G Oliver
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, 27708, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Kwon-Sik Park
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
24
|
Duplaquet L, So K, Ying AW, Li X, Li Y, Qiu X, Li R, Singh S, Wu XS, Liu Q, Qi J, Somerville TDD, Heiling H, Mazzola E, Lee Y, Zoller T, Vakoc CR, Doench JG, Forrester WC, Abrams T, Long HW, Niederst MJ, Kadoch C, Oser MG. Mammalian SWI/SNF complex activity regulates POU2F3 and constitutes a targetable dependency in small cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.21.576304. [PMID: 38328215 PMCID: PMC10849479 DOI: 10.1101/2024.01.21.576304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Small cell lung cancers (SCLC) are comprised of heterogeneous subtypes marked by lineage-specific transcription factors, including ASCL1, NEUROD1, and POU2F3. POU2F3-positive SCLC, ∼12% of all cases, are uniquely dependent on POU2F3 itself; as such, approaches to attenuate POU2F3 expression may represent new therapeutic opportunities. Here using genome-scale screens for regulators of POU2F3 expression and SCLC proliferation, we define mSWI/SNF complexes, including non-canonical BAF (ncBAF) complexes, as top dependencies specific to POU2F3-positive SCLC. Notably, clinical-grade pharmacologic mSWI/SNF inhibition attenuates proliferation of all POU2F3-positive SCLCs, while disruption of ncBAF via BRD9 degradation is uniquely effective in pure non-neuroendocrine POU2F3-SCLCs. mSWI/SNF maintains accessibility over gene loci central to POU2F3-mediated gene regulatory networks. Finally, chemical targeting of SMARCA4/2 mSWI/SNF ATPases and BRD9 decrease POU2F3-SCLC tumor growth and increase survival in vivo . Taken together, these results characterize mSWI/SNF-mediated global governance of the POU2F3 oncogenic program and suggest mSWI/SNF inhibition as a therapeutic strategy for SCLC.
Collapse
|
25
|
Vocino Trucco G, Righi L, Volante M, Papotti M. Updates on lung neuroendocrine neoplasm classification. Histopathology 2024; 84:67-85. [PMID: 37794655 DOI: 10.1111/his.15058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023]
Abstract
Lung neuroendocrine neoplasms (NENs) are a heterogeneous group of pulmonary neoplasms showing different morphological patterns and clinical and biological characteristics. The World Health Organisation (WHO) classification of lung NENs has been recently updated as part of the broader attempt to uniform the classification of NENs. This much-needed update has come at a time when insights from seminal molecular characterisation studies revolutionised our understanding of the biological and pathological architecture of lung NENs, paving the way for the development of novel diagnostic techniques, prognostic factors and therapeutic approaches. In this challenging and rapidly evolving landscape, the relevance of the 2021 WHO classification has been recently questioned, particularly in terms of its morphology-orientated approach and its prognostic implications. Here, we provide a state-of-the-art review on the contemporary understanding of pulmonary NEN morphology and the potential contribution of artificial intelligence, the advances in NEN molecular profiling with their impact on the classification system and, finally, the key current and upcoming prognostic factors.
Collapse
Affiliation(s)
| | - Luisella Righi
- Department of Oncology, University of Turin, Turin, Italy
| | - Marco Volante
- Department of Oncology, University of Turin, Turin, Italy
| | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
26
|
Berezowska S, Maillard M, Keyter M, Bisig B. Pulmonary squamous cell carcinoma and lymphoepithelial carcinoma - morphology, molecular characteristics and differential diagnosis. Histopathology 2024; 84:32-49. [PMID: 37936498 DOI: 10.1111/his.15076] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 11/09/2023]
Abstract
Squamous cell carcinoma (SCC) comprises one of the major groups of non-small-cell carcinoma of the lung, and is subtyped into keratinising, non-keratinising and basaloid SCC. SCC can readily be diagnosed using histomorphology alone in keratinising SCC. Confirmatory immunohistochemical analyses should always be applied in non-keratinising and basaloid tumours to exclude differential diagnoses, most prominently adenocarcinoma and high-grade neuroendocrine carcinoma, which may have important therapeutic consequences. According to the World Health Organisation (WHO) classification 2015, the diagnosis of SCC can be rendered in resections of morphologically ambiguous tumours with squamous immunophenotype. In biopsies and cytology preparations in the same setting the current guidelines propose a diagnosis of 'non-small-cell carcinoma, favour SCC' in TTF1-negative and p40-positive tumours to acknowledge a possible sampling bias and restrict extended immunohistochemical evaluation in order to preserve tissue for molecular testing. Most SCC feature a molecular 'tobacco-smoke signature' with enrichment in GG > TT mutations, in line with the strong epidemiological association of SCC with smoking. Targetable mutations are extremely rare but they do occur, in particular in younger and non- or light-smoking patients, warranting molecular investigations. Lymphoepithelial carcinoma (LEC) is a poorly differentiated SCC with a syncytial growth pattern and a usually prominent lymphoplasmacytic infiltrate and frequent Epstein-Barr virus (EBV) association. In this review, we describe the morphological and molecular characteristics of SCC and LEC and discuss the most pertinent differential diagnoses.
Collapse
Affiliation(s)
- Sabina Berezowska
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Marie Maillard
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Mark Keyter
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Bettina Bisig
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
27
|
Jimbo N, Ohbayashi C, Takeda M, Fujii T, Mitsui S, Tsukamoto R, Tanaka Y, Itoh T, Maniwa Y. POU2F3-Expressing Small Cell Lung Carcinoma and Large Cell Neuroendocrine Carcinoma Show Morphologic and Phenotypic Overlap. Am J Surg Pathol 2024; 48:4-15. [PMID: 37904277 DOI: 10.1097/pas.0000000000002145] [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: 11/01/2023]
Abstract
Considering the differences in protein expression in small cell lung carcinoma (SCLC) by molecular classification, it is likely that there are differences in morphology, but the relationship between molecular classification and morphology has not been examined. Furthermore, there are limited reports concerning this molecular classification for large cell neuroendocrine carcinoma (LCNEC) and SCLC simultaneously. Therefore, we investigated the relationship between immunohistochemistry-based molecular classification and morphology, protein expression, and clinical features of 146 consecutive resection specimens of pulmonary neuroendocrine carcinoma (NEC), focusing mainly on POU2F3, the master transcription factor involved in tuft cell generation. POU2F3-dominant SCLC (n=24) and LCNEC (n=14) showed overlap in cytomorphology, while non-POU2F3-dominant SCLC (n=71) and LCNEC (n=37) showed distinct differences in cytomorphology. In addition, POU2F3-dominant NEC exhibited significantly more abundant tumor stroma, more prominent nest formation, more frequent bronchial intraepithelial involvement, and less frequent background fibrosis than non-POU2F3-dominant NEC. Immunohistochemically, POU2F3-dominant SCLC and LCNEC were characterized by lower expression of TTF-1, CEA, and neuroendocrine markers and higher expression of bcl-2, c-Myc, and c-kit. Clinically, POU2F3-dominant NEC had a significantly better prognosis than non-POU2F3-dominant NEC for recurrence-free survival. POU2F3-dominant NEC had a higher smoking index than non-POU2F3-dominant NEC. POU2F3-dominant NEC forms a unique population, exhibiting intermediate morphologic features between SCLC and LCNEC, with distinct protein expression as tuft cell-like carcinoma. Recognition of this unique subtype may provide clues for solving the long-standing issues of NEC and appropriate therapeutic stratification. It is important to accurately identify POU2F3-expressing carcinomas by immunohistochemistry and to analyze their clinicopathological features.
Collapse
Affiliation(s)
- Naoe Jimbo
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Chiho Ohbayashi
- Department of Diagnostic Pathology, Shinko Hospital, Chuo-ku, Kobe, Japan
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara, Japan, Kashihara, Nara, Japan
| | - Maiko Takeda
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara, Japan, Kashihara, Nara, Japan
| | - Tomomi Fujii
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara, Japan, Kashihara, Nara, Japan
| | - Suguru Mitsui
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Ryuko Tsukamoto
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Yugo Tanaka
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Tomoo Itoh
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Yoshimasa Maniwa
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| |
Collapse
|
28
|
Ng J, Pan E, Johnston A, Ribera NT, Kersbergen A, Hess JB, Best SA, Tsui E, Steinfort D, Sutherland KD. A Multiplexed Approach to Assess Small Cell Lung Cancer Subtype Heterogeneity in Primary and Patient-Derived Tumor Samples. Methods Mol Biol 2024; 2806:117-138. [PMID: 38676800 DOI: 10.1007/978-1-0716-3858-3_10] [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: 04/29/2024]
Abstract
Unlocking the heterogeneity of cancers is crucial for developing therapeutic approaches that effectively eradicate disease. As our understanding of markers specific to cancer subclones or subtypes expands, there is a growing demand for advanced technologies that enable the simultaneous investigation of multiple targets within an individual tumor sample. Indeed, multiplex approaches offer distinct benefits, particularly when tumor specimens are small and scarce. Here we describe the utility of two fluorescence-based multiplex approaches; fluorescent Western blots, and multiplex immunohistochemistry (Opal™) staining to interrogate heterogeneity, using small cell lung cancer as an example. Critically, the coupling of Opal™ staining with advanced image quantitation, permits the dissection of cancer cell phenotypes at a single cell level. These approaches can be applied to patient biopsies and/or patient-derived xenograft (PDX) models and serve as powerful methodologies for assessing tumor cell heterogeneity in response to therapy or between metastatic lesions across diverse tissue sites.
Collapse
Affiliation(s)
- Jin Ng
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Emma Pan
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Alex Johnston
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Nina Tubau Ribera
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Ariena Kersbergen
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jonas B Hess
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Sarah A Best
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Ellen Tsui
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Daniel Steinfort
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
- Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Kate D Sutherland
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
29
|
Denize T, Meador CB, Rider AB, Ganci ML, Barth JL, Kem M, Mino-Kenudson M, Hung YP. Concordance of ASCL1, NEUROD1 and POU2F3 transcription factor-based subtype assignment in paired tumour samples from small cell lung carcinoma. Histopathology 2023; 83:912-924. [PMID: 37644667 DOI: 10.1111/his.15034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/27/2023] [Accepted: 08/10/2023] [Indexed: 08/31/2023]
Abstract
AIMS Small cell lung carcinoma (SCLC) can be classified into transcription factor-based subtypes (ASCL1, NeuroD1, POU2F3). While in-vitro studies suggest intratumoral heterogeneity in the expression of these markers, how SCLC subtypes vary over time and among locations in patients remains unclear. METHODS AND RESULTS We searched a consecutive series of patients at our institution in 2006-22 for those with greater than one available formalin-fixed paraffin-embedded SCLC sample in multiple sites and/or time-points. Immunohistochemistry for ASCL1, NeuroD1 and POU2F3 was performed and evaluated using H-scores, with subtype assigned based on the positive marker (H-score threshold >10) with the highest H-score. The 179 samples (75, lung; 51, lymph nodes; 53, non-nodal metastases) from 84 patients (74 with two, 10 with more than two samples) included 98 (54.7%) ASCL1-dominant, 47 (26.3%) NeuroD1-dominant, 15 (8.4%) POU2F3-dominant, 17 (9.5%) triple-negative and two (1.1%) ASCL1/NeuroD1 co-dominant samples. NeuroD1-dominant subtype was enriched in non-lung locations. Subtype concordance from pairwise comparison was 71.4% overall and 89.7% after accounting for ASCL1/NeuroD1-dual expressors and technical factors including <500 cells/slide, H-score thresholds and sample decalcification. No significant difference in subtype concordance was noted with a longer time lapse or with extrathoracic versus intrathoracic samples in this cohort. CONCLUSIONS After accounting for technical factors, transcription factor-based subtyping was discordant among multiple SCLC samples in ~10% of patients, regardless of sample locations and time lapse. Our findings highlighted the spatiotemporal heterogeneity of SCLC in clinical samples and potential challenges, including technical and biological factors, that might limit concordance in SCLC transcription factor-based subtyping.
Collapse
Affiliation(s)
- Thomas Denize
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Catherine B Meador
- Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Anna B Rider
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Maria L Ganci
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Jaimie L Barth
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Marina Kem
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Yin P Hung
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
30
|
Megyesfalvi Z, Gay CM, Popper H, Pirker R, Ostoros G, Heeke S, Lang C, Hoetzenecker K, Schwendenwein A, Boettiger K, Bunn PA, Renyi-Vamos F, Schelch K, Prosch H, Byers LA, Hirsch FR, Dome B. Clinical insights into small cell lung cancer: Tumor heterogeneity, diagnosis, therapy, and future directions. CA Cancer J Clin 2023; 73:620-652. [PMID: 37329269 DOI: 10.3322/caac.21785] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023] Open
Abstract
Small cell lung cancer (SCLC) is characterized by rapid growth and high metastatic capacity. It has strong epidemiologic and biologic links to tobacco carcinogens. Although the majority of SCLCs exhibit neuroendocrine features, an important subset of tumors lacks these properties. Genomic profiling of SCLC reveals genetic instability, almost universal inactivation of the tumor suppressor genes TP53 and RB1, and a high mutation burden. Because of early metastasis, only a small fraction of patients are amenable to curative-intent lung resection, and these individuals require adjuvant platinum-etoposide chemotherapy. Therefore, the vast majority of patients are currently being treated with chemoradiation with or without immunotherapy. In patients with disease confined to the chest, standard therapy includes thoracic radiotherapy and concurrent platinum-etoposide chemotherapy. Patients with metastatic (extensive-stage) disease are treated with a combination of platinum-etoposide chemotherapy plus immunotherapy with an anti-programmed death-ligand 1 monoclonal antibody. Although SCLC is initially very responsive to platinum-based chemotherapy, these responses are transient because of the development of drug resistance. In recent years, the authors have witnessed an accelerating pace of biologic insights into the disease, leading to the redefinition of the SCLC classification scheme. This emerging knowledge of SCLC molecular subtypes has the potential to define unique therapeutic vulnerabilities. Synthesizing these new discoveries with the current knowledge of SCLC biology and clinical management may lead to unprecedented advances in SCLC patient care. Here, the authors present an overview of multimodal clinical approaches in SCLC, with a special focus on illuminating how recent advancements in SCLC research could accelerate clinical development.
Collapse
Affiliation(s)
- Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Helmut Popper
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Robert Pirker
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Gyula Ostoros
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Schwendenwein
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Paul A Bunn
- University of Colorado School of Medicine, Aurora, CO, USA
| | - Ferenc Renyi-Vamos
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Helmut Prosch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Lauren A Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Tisch Cancer Institute, Center for Thoracic Oncology, Mount Sinai Health System, New York, NY, USA
| | - Balazs Dome
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Translational Medicine, Lund University, Lund, Sweden
| |
Collapse
|
31
|
Zhang SL, Zhang CY, Chen YQ, Li YF, Xie Z, Zhang XC, Zhou Q, Zhong WZ, Huang J, Sun H, Zheng MY, Xiao FM, Yan HH, Lu DX, Lv ZY, Wu YL, Chen HJ, Yang JJ. Expression of EGFR-mutant proteins and genomic evolution in EGFR-mutant transformed small cell lung cancer. J Thorac Dis 2023; 15:4620-4635. [PMID: 37868836 PMCID: PMC10586967 DOI: 10.21037/jtd-23-161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/28/2023] [Indexed: 10/24/2023]
Abstract
Background The transformation of epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma (LUAD) into small cell lung cancer (SCLC) accounts for 3-14% of the resistance mechanism to EGFR tyrosine kinase inhibitors (TKIs). At present, there is no relevant research to explore the dynamic expression of EGFR-mutant proteins and genomic evolution in EGFR-mutant transformed SCLC/neuroendocrine carcinoma (NEC). Methods Genetic analysis and protein level analysis by next-generation sequencing (NGS), Whole-exome sequencing (WES) and immunohistochemistry were performed to explore expression of EGFR-mutant proteins and genomic evolution in EGFR-mutant transformed SCLC. The research used three patient-derived organoids (PDOs) to explore the efficacy of combo [chemotherapy (chemo) plus TKI or bevacizumab] treatment. According to the subsequent treatment regimens after SCLC/NEC transformation, 35 patients were divided into chemo (n=21) and combo (n=14) groups. Results EGFR L858R and EGFR E746-750 del protein expression by immunohistochemistry was 80.0% (4/5) and 100% (6/6), respectively (P=0.455) in initially-transformed tissues. Meanwhile, EGFR-mutant proteins were expressed in 85.7% (6/7) of dynamic rebiopsy tissues or effusion samples after the first transformation. Then, by the pathway enrichment analysis of tissue and plasma NGS, the EGFR-related pathways were still activated after SCLC/NEC transformation. Moreover, WES analysis revealed that transformed SCLC shared a common clonal origin from the baseline LUAD. The drug sensitivity of three PDOs demonstrated potent anti-cancer activity of EGFR-TKIs plus chemo, compared with chemo or TKI alone. There were significant differences in objective response rate (ORR) between the combo and chemo groups [42.9 % vs. 4.8%, P=0.010, 95% confidence interval (CI): 1.5-145.2]. Furthermore, the median post-transformation progression-free survival (pPFS) was significantly prolonged in the combo group, with 5.4 (95% CI: 3.4-7.4) versus 3.5 (95% CI: 2.7-4.3, P=0.012) months. Conclusions EGFR 19del or L858R-mutant proteins could be constantly expressed, and EGFR pathway still existed in EGFR-mutant transformed SCLC/NEC with a common clonal origin from the baseline LUAD. Taking together, these molecular characteristics potentially favored clinical efficacy in transformed SCLC/NEC treated with the combo regimen.
Collapse
Affiliation(s)
- Shi-Ling Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chan-Yuan Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu-Qing Chen
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu-Fa Li
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhi Xie
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jie Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hao Sun
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ming-Ying Zheng
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fa-Man Xiao
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hong-Hong Yan
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dan-Xia Lu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhi-Yi Lv
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hua-Jun Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
32
|
Wang Y, Jin Y, Shen X, Zheng Q, Xue Q, Chen L, Lin Y, Li Y. POU2F3: A Sensitive and Specific Diagnostic Marker for Neuroendocrine-low/negative Small Cell Lung Cancer. Am J Surg Pathol 2023; 47:1059-1066. [PMID: 37357936 DOI: 10.1097/pas.0000000000002081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
POU2F3 (POU class 2 homeobox 3) is a novel transcription factor used to define the special molecular subtype of small cell lung cancer (SCLC) known as SCLC-P. Nevertheless, the sensitivity and specificity of POU2F3 immunohistochemical (IHC) staining have not been fully investigated. In this study, we explored the expression of POU2F3 by IHC in a large cohort of SCLC clinical samples (n=246), other common lung cancer types (n=2207), and various other cancer types (n=194). The results showed that POU2F3 was strongly nuclear stained in 13.41% (33/246) of SCLC cases, with negative or minimal labeling for thyroid transcription factor-1 and neuroendocrine (NE) markers. Compared with POU2F3-negative SCLC, SCLC-P harbored fewer TP53 and RB1 mutations. POU2F3 was also expressed in 3.13% (8/256) of squamous cell carcinomas (SCCs) and 20% (2/10) of large cell NE carcinomas (LCNECs), whereas other lung cancer types were negative. In addition to lung cancer, POU2F3 was positive in 22.2% (4/18) of thymic tumors. All other tumors were POU2F3-negative except for thymic carcinoma, although sparsely distributed weak nuclear staining was observed in lung adenocarcinoma, cervical SCC, and colorectal carcinoma. The sensitivity and specificity of POU2F3 in NE-low/negative SCLC were 82.1% and 99.4%, respectively. Notably, some rare unique patterns of POU2F3 expression were observed. One case of thymic SCC was characterized by diffuse and uniform cytomembrane staining. One case of esophageal NE tumor was nuclear-positive, while the normal proliferating squamous epithelium was strongly membrane-stained. This is the largest cohort of clinical samples to confirm that POU2F3 is a highly sensitive and specific diagnostic marker for NE-low/negative SCLC.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Jin
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuxia Shen
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Zheng
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qianqian Xue
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lijun Chen
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yicong Lin
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Li
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Duplaquet L, Li Y, Booker MA, Xie Y, Olsen SN, Patel RA, Hong D, Hatton C, Denize T, Walton E, Laimon YN, Li R, Jiang Y, Bronson RT, Southard J, Li S, Signoretti S, Qiu X, Cejas P, Armstrong SA, Long HW, Tolstorukov MY, Haffner MC, Oser MG. KDM6A epigenetically regulates subtype plasticity in small cell lung cancer. Nat Cell Biol 2023; 25:1346-1358. [PMID: 37591951 PMCID: PMC10546329 DOI: 10.1038/s41556-023-01210-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Small cell lung cancer (SCLC) exists broadly in four molecular subtypes: ASCL1, NEUROD1, POU2F3 and Inflammatory. Initially, SCLC subtypes were thought to be mutually exclusive, but recent evidence shows intra-tumoural subtype heterogeneity and plasticity between subtypes. Here, using a CRISPR-based autochthonous SCLC genetically engineered mouse model to study the consequences of KDM6A/UTX inactivation, we show that KDM6A inactivation induced plasticity from ASCL1 to NEUROD1 resulting in SCLC tumours that express both ASCL1 and NEUROD1. Mechanistically, KDM6A normally maintains an active chromatin state that favours the ASCL1 subtype with its loss decreasing H3K4me1 and increasing H3K27me3 at enhancers of neuroendocrine genes leading to a cell state that is primed for ASCL1-to-NEUROD1 subtype switching. This work identifies KDM6A as an epigenetic regulator that controls ASCL1 to NEUROD1 subtype plasticity and provides an autochthonous SCLC genetically engineered mouse model to model ASCL1 and NEUROD1 subtype heterogeneity and plasticity, which is found in 35-40% of human SCLCs.
Collapse
Affiliation(s)
- Leslie Duplaquet
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yixiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew A Booker
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yingtian Xie
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sarah Naomi Olsen
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
| | - Radhika A Patel
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Deli Hong
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Charlie Hatton
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
| | - Thomas Denize
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily Walton
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yasmin N Laimon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rong Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yijia Jiang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Roderick T Bronson
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Jackson Southard
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuqiang Li
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xintao Qiu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paloma Cejas
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott A Armstrong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Y Tolstorukov
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael C Haffner
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Matthew G Oser
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, MA, USA.
| |
Collapse
|
34
|
Lo YC, Rivera-Concepcion J, Vasmatzis G, Aubry MC, Leventakos K. Subtype of SCLC Is an Intrinsic and Persistent Feature Through Systemic Treatment. JTO Clin Res Rep 2023; 4:100561. [PMID: 37731627 PMCID: PMC10507151 DOI: 10.1016/j.jtocrr.2023.100561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/29/2023] [Accepted: 08/06/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction SCLC is an aggressive malignancy with poor outcome. Most patients have disease recurrence despite treatments with multiple modalities. Subtyping of SCLC has been proposed recently, and novel agents targeting specific subtypes are actively being investigated. In this study, we evaluated the plasticity of subtypes in paired pre- and post-treatment samples. The aim was to understand possible subtype evolution after chemotherapy resistance that could lead to alternate targeted therapy strategies. Methods A total of 68 samples from 32 patients with sufficient paired specimens were identified from 1998 to 2022. ASCL1, NEUROD1, and POU2F3 immunohistochemistry studies were performed on all cases, and subtyping by predominant expression was determined. Subtype comparison in each patient was performed, and expression analysis was performed on the basis of subtypes. Results Of 32 cases, 28 (88%) had the same subtype in pre- and first post-treatment specimens. Protein expression level of subtype-specific transcription factor remained stable after chemotherapy. Two of five (40%) NEUROD1-predominant SCLC switched to ASCL1-predominant phenotype after treatment. One case had a pitfall of scoring ASCL1 on specimen with marked crushing artifacts. One case revealed the challenge of proper subtyping for samples with borderline POU2F3 expression. Conclusions Subtype of SCLC generally remains the same after acquiring chemotherapy resistance. Plasticity was observed with rare cases switching from NEUROD1-predominant to ASC1-predominant SCLC. Resubtyping is unnecessary for the consideration of novel subtype-specific targeted agents, except cases with NEUROD1-predominant subtype.
Collapse
Affiliation(s)
- Ying-Chun Lo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Joel Rivera-Concepcion
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
- Current Affiliation: Department of Medical Oncology, Duke Cancer Center, Durham, North Carolina
| | - George Vasmatzis
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | | |
Collapse
|
35
|
Wei W, Zhang W, Wu S, Duan W, Wang Z. Advances in tuft cells, a chemosensory cell in sequential diseases of the pancreas. Biochim Biophys Acta Rev Cancer 2023; 1878:188911. [PMID: 37182665 DOI: 10.1016/j.bbcan.2023.188911] [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: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
Tuft cells are solitary chemosensory cells distributed mainly in hollow organs and detected in human and mouse pancreas precursor lesions of pancreatic cancer. Induced by inflammation and KRAS mutation, pancreatic acinar cell-derived tuft cells play a protective role in epithelium injury. The tumour suppression of tuft cells has been indicated in some studies. However, the function of tuft cells in pancreatic cancer remains unclear. In this review, we first introduce the definition of tuft cells and then review the relationship between tuft cells and pancreatic inflammation. In addition, we emphasized the role of tuft cells in the genesis and development of pancreatic cancers, especially the part of markers for tuft cell's doublecortin-like kinase 1 (DCLK1). Finally, we turn to the microscopic perspective and review the interactions between tuft cells and the microbiome in the pancreatic microenvironment. Overall, we describe the role of tuft cells in response to tissue damage and tumour progression in the pancreas. Nevertheless, the specific formation principle and the more detailed mechanism of action of tuft cells in the pancreas remain to be further explored.
Collapse
Affiliation(s)
- Wanzhen Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Weifan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Shuai Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China.
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China.
| |
Collapse
|
36
|
Yamada Y. Histogenetic and disease-relevant phenotypes in thymic epithelial tumors (TETs): The potential significance for future TET classification. Pathol Int 2023; 73:265-280. [PMID: 37278579 DOI: 10.1111/pin.13343] [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: 03/06/2023] [Accepted: 05/18/2023] [Indexed: 06/07/2023]
Abstract
Thymic epithelial tumors (TETs) encompass morphologically various subtypes. Thus, it would be meaningful to explore the expression phenotypes that delineate each TET subtype or overarching multiple subtypes. If these profiles are related to thymic physiology, they will improve our biological understanding of TETs and may contribute to the establishment of a more rational TET classification. Against this background, pathologists have attempted to identify histogenetic features in TETs for a long time. As part of this work, our group has reported several TET expression profiles that are histotype-dependent and related to the nature of thymic epithelial cells (TECs). For example, we found that beta5t, a constituent of thymoproteasome unique to cortical TECs, is expressed mainly in type B thymomas, for which the nomenclature of cortical thymoma was once considered. Another example is the discovery that most thymic carcinomas, especially thymic squamous cell carcinomas, exhibit expression profiles similar to tuft cells, a recently discovered special type of medullary TEC. This review outlines the currently reported histogenetic phenotypes of TETs, including those related to thymoma-associated myasthenia gravis, summarizes their genetic signatures, and provides a perspective for the future direction of TET classification.
Collapse
Affiliation(s)
- Yosuke Yamada
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
37
|
Naso JR, Vrana JA, Koepplin JW, Molina JR, Roden AC. EZH2 and POU2F3 Can Aid in the Distinction of Thymic Carcinoma from Thymoma. Cancers (Basel) 2023; 15:cancers15082274. [PMID: 37190202 DOI: 10.3390/cancers15082274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Thymic carcinoma is an aggressive malignancy that can be challenging to distinguish from thymoma using histomorphology. We assessed two emerging markers for these entities, EZH2 and POU2F3, and compared them with conventional immunostains. Whole slide sections of 37 thymic carcinomas, 23 type A thymomas, 13 type B3 thymomas, and 8 micronodular thymomas with lymphoid stroma (MNTLS) were immunostained for EZH2, POU2F3, CD117, CD5, TdT, BAP1, and MTAP. POU2F3 (≥10% hotspot staining), CD117, and CD5 showed 100% specificity for thymic carcinoma versus thymoma with 51%, 86%, and 35% sensitivity, respectively, for thymic carcinoma. All POU2F3 positive cases were also positive for CD117. All thymic carcinomas showed >10% EZH2 staining. EZH2 (≥80% staining) had a sensitivity of 81% for thymic carcinoma and a specificity of 100% for thymic carcinoma versus type A thymoma and MNTLS but had poor specificity (46%) for thymic carcinoma versus B3 thymoma. Adding EZH2 to a panel of CD117, TdT, BAP1, and MTAP increased cases with informative results from 67/81 (83%) to 77/81 (95%). Overall, absent EZH2 staining may be useful for excluding thymic carcinoma, diffuse EZH2 staining may help to exclude type A thymoma and MNTLS, and ≥10% POU2F3 staining has excellent specificity for thymic carcinoma versus thymoma.
Collapse
Affiliation(s)
- Julia R Naso
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
| | - Julie A Vrana
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
| | - Justin W Koepplin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
| | - Julian R Molina
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55902, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
38
|
Neuroendocrine neoplasms of the lung and gastrointestinal system: convergent biology and a path to better therapies. Nat Rev Clin Oncol 2023; 20:16-32. [PMID: 36307533 DOI: 10.1038/s41571-022-00696-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2022] [Indexed: 11/08/2022]
Abstract
Neuroendocrine neoplasms (NENs) can develop in almost any organ and span a spectrum from well-differentiated and indolent neuroendocrine tumours (NETs) to poorly differentiated and highly aggressive neuroendocrine carcinomas (NECs), including small-cell lung cancer (SCLC). These neoplasms are thought to primarily derive from neuroendocrine precursor cells located throughout the body and can also arise through neuroendocrine transdifferentiation of organ-specific epithelial cell types. Hence, NENs constitute a group of tumour types that share key genomic and phenotypic characteristics irrespective of their site of origin, albeit with some organ-specific differences. The establishment of representative preclinical models for several of these disease entities together with analyses of human tumour specimens has provided important insights into crucial aspects of their biology with therapeutic implications. In this Review, we provide a comprehensive overview of the current understanding of NENs of the gastrointestinal system and lung from clinical and biological perspectives. Research on NENs has typically been siloed by the tumour site of origin, and a cross-cutting view might enable advances in one area to accelerate research in others. Therefore, we aim to emphasize that a better understanding of the commonalities and differences of NENs arising in different organs might more effectively inform clinical research to define therapeutic targets and ultimately optimize patient care.
Collapse
|
39
|
Pulmonary cancers across different histotypes share hybrid tuft cell/ionocyte-like molecular features and potentially druggable vulnerabilities. Cell Death Dis 2022; 13:979. [PMID: 36402755 PMCID: PMC9675833 DOI: 10.1038/s41419-022-05428-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022]
Abstract
Tuft cells are chemosensory epithelial cells in the respiratory tract and several other organs. Recent studies revealed tuft cell-like gene expression signatures in some pulmonary adenocarcinomas, squamous cell carcinomas (SQCC), small cell carcinomas (SCLC), and large cell neuroendocrine carcinomas (LCNEC). Identification of their similarities could inform shared druggable vulnerabilities. Clinicopathological features of tuft cell-like (tcl) subsets in various lung cancer histotypes were studied in two independent tumor cohorts using immunohistochemistry (n = 674 and 70). Findings were confirmed, and additional characteristics were explored using public datasets (RNA seq and immunohistochemical data) (n = 555). Drug susceptibilities of tuft cell-like SCLC cell lines were also investigated. By immunohistochemistry, 10-20% of SCLC and LCNEC, and approximately 2% of SQCC expressed POU2F3, the master regulator of tuft cells. These tuft cell-like tumors exhibited "lineage ambiguity" as they co-expressed NCAM1, a marker for neuroendocrine differentiation, and KRT5, a marker for squamous differentiation. In addition, tuft cell-like tumors co-expressed BCL2 and KIT, and tuft cell-like SCLC and LCNEC, but not SQCC, also highly expressed MYC. Data from public datasets confirmed these features and revealed that tuft cell-like SCLC and LCNEC co-clustered on hierarchical clustering. Furthermore, only tuft cell-like subsets among pulmonary cancers significantly expressed FOXI1, the master regulator of ionocytes, suggesting their bidirectional but immature differentiation status. Clinically, tuft cell-like SCLC and LCNEC had a similar prognosis. Experimentally, tuft cell-like SCLC cell lines were susceptible to PARP and BCL2 co-inhibition, indicating synergistic effects. Taken together, pulmonary tuft cell-like cancers maintain histotype-related clinicopathologic characteristics despite overlapping unique molecular features. From a therapeutic perspective, identification of tuft cell-like LCNECs might be crucial given their close kinship with tuft cell-like SCLC.
Collapse
|
40
|
Yamada Y, Bohnenberger H, Kriegsmann M, Kriegsmann K, Sinn P, Goto N, Nakanishi Y, Seno H, Chigusa Y, Fujimoto M, Minamiguchi S, Haga H, Simon R, Sauter G, Ströbel P, Marx A. Tuft cell-like carcinomas: novel cancer subsets present in multiple organs sharing a unique gene expression signature. Br J Cancer 2022; 127:1876-1885. [PMID: 35999270 PMCID: PMC9643388 DOI: 10.1038/s41416-022-01957-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Tuft cells are chemosensory epithelial cells playing a role in innate immunity. Recent studies revealed cancers with a tuft cell-like gene expression signature in the thorax. We wondered whether this signature might also occur in extrathoracic cancers. METHODS We examined mRNA expression of tuft cell markers (POU2F3, GFI1B, TRPM5, SOX9, CHAT, and AVIL) in 19 different types of cancers in multiple extrathoracic organs with The Cancer Genome Atlas (TCGA) (N = 6322). Four different extrathoracic cancers in our local archives (N = 909) were analysed by immunohistochemistry. RESULTS Twenty-two (0.35%) extrathoracic tumours with co-expression of POU2F3 and other tuft cell markers were identified in various TCGA datasets. Twelve of the 22 "tuft cell-like tumours" shared poor differentiation and a gene expression pattern, including KIT, anti-apoptotic BCL2, and ionocyte-associated genes. In our archival cases, eleven (1.21%) tumours co-expressing POU2F3, KIT, and BCL2 on immunohistochemistry, i.e., were presumable tuft cell-like cancers. In three among five TCGA cohorts, the tuft cell-like cancer subsets expressed SLFN11, a promising biomarker of PARP inhibitor susceptibility. CONCLUSIONS Tuft cell-like carcinomas form distinct subsets in cancers of many organs. It appears warranted to investigate their shared gene expression signature as a predictive biomarker for novel therapeutic strategies.
Collapse
Affiliation(s)
- Yosuke Yamada
- Institute of Pathology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan.
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Lung Cancer Research (DZL), Heidelberg, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Norihiro Goto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | - Hironori Haga
- Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
41
|
Szeitz B, Megyesfalvi Z, Woldmar N, Valkó Z, Schwendenwein A, Bárány N, Paku S, László V, Kiss H, Bugyik E, Lang C, Szász AM, Pizzatti L, Bogos K, Hoda MA, Hoetzenecker K, Marko-Varga G, Horvatovich P, Döme B, Schelch K, Rezeli M. In-depth proteomic analysis reveals unique subtype-specific signatures in human small-cell lung cancer. Clin Transl Med 2022; 12:e1060. [PMID: 36149789 PMCID: PMC9506422 DOI: 10.1002/ctm2.1060] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 11/12/2022] Open
Abstract
Background Small‐cell lung cancer (SCLC) molecular subtypes have been primarily characterized based on the expression pattern of the following key transcription regulators: ASCL1 (SCLC‐A), NEUROD1 (SCLC‐N), POU2F3 (SCLC‐P) and YAP1 (SCLC‐Y). Here, we investigated the proteomic landscape of these molecular subsets with the aim to identify novel subtype‐specific proteins of diagnostic and therapeutic relevance. Methods Pellets and cell media of 26 human SCLC cell lines were subjected to label‐free shotgun proteomics for large‐scale protein identification and quantitation, followed by in‐depth bioinformatic analyses. Proteomic data were correlated with the cell lines’ phenotypic characteristics and with public transcriptomic data of SCLC cell lines and tissues. Results Our quantitative proteomic data highlighted that four molecular subtypes are clearly distinguishable at the protein level. The cell lines exhibited diverse neuroendocrine and epithelial–mesenchymal characteristics that varied by subtype. A total of 367 proteins were identified in the cell pellet and 34 in the culture media that showed significant up‐ or downregulation in one subtype, including known druggable proteins and potential blood‐based markers. Pathway enrichment analysis and parallel investigation of transcriptomics from SCLC cell lines outlined unique signatures for each subtype, such as upregulated oxidative phosphorylation in SCLC‐A, DNA replication in SCLC‐N, neurotrophin signalling in SCLC‐P and epithelial–mesenchymal transition in SCLC‐Y. Importantly, we identified the YAP1‐driven subtype as the most distinct SCLC subgroup. Using sparse partial least squares discriminant analysis, we identified proteins that clearly distinguish four SCLC subtypes based on their expression pattern, including potential diagnostic markers for SCLC‐Y (e.g. GPX8, PKD2 and UFO). Conclusions We report for the first time, the protein expression differences among SCLC subtypes. By shedding light on potential subtype‐specific therapeutic vulnerabilities and diagnostic biomarkers, our results may contribute to a better understanding of SCLC biology and the development of novel therapies.
Collapse
Affiliation(s)
- Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Zsolt Megyesfalvi
- National Korányi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Budapest, Hungary
| | - Nicole Woldmar
- Division of Clinical Protein Science, & Imaging, Department of Clinical Sciences (Lund) and Department of Biomedical Engineering, Lund University, Lund, Sweden.,Laboratory of Molecular Biology and Proteomics of Blood/LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Zsuzsanna Valkó
- National Korányi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Anna Schwendenwein
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Nándor Bárány
- National Korányi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Sándor Paku
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Viktória László
- National Korányi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Helga Kiss
- Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Budapest, Hungary.,University of Pécs, Pécs, Hungary
| | - Edina Bugyik
- National Korányi Institute of Pulmonology, Budapest, Hungary.,First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Christian Lang
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Attila Marcell Szász
- National Korányi Institute of Pulmonology, Budapest, Hungary.,Department of Bioinformatics, Semmelweis University, Budapest, Hungary
| | - Luciana Pizzatti
- Laboratory of Molecular Biology and Proteomics of Blood/LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Krisztina Bogos
- National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - György Marko-Varga
- Division of Clinical Protein Science, & Imaging, Department of Clinical Sciences (Lund) and Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Balázs Döme
- National Korányi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Budapest, Hungary.,Department of Translational Medicine, Lund University, Lund, Sweden
| | - Karin Schelch
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Melinda Rezeli
- Division of Clinical Protein Science, & Imaging, Department of Clinical Sciences (Lund) and Department of Biomedical Engineering, Lund University, Lund, Sweden
| |
Collapse
|
42
|
Determining Whether YAP1 and POU2F3 Are Antineuroendocrine Factors. J Thorac Oncol 2022; 17:1070-1073. [PMID: 36031286 DOI: 10.1016/j.jtho.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 11/22/2022]
|
43
|
Keogh A, Finn S, Radonic T. Emerging Biomarkers and the Changing Landscape of Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14153772. [PMID: 35954436 PMCID: PMC9367597 DOI: 10.3390/cancers14153772] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Small cell lung cancer (SCLC) is an aggressive cancer representing 15% of all lung cancers. Unlike other types of lung cancer, treatments for SCLC have changed very little in the past 20 years and therefore, the survival rate remains low. This is due, in part, to the lack of understanding of the biological basis of this disease and the previous idea that all SCLCs are the same. Multiple recent studies have identified that SCLCs have varying biological activity and can be divided into four different groups. The advantage of this is that each of these four groups responds differently to new treatments, which hopefully will dramatically improve survival. Additionally, the aim of these new treatments is to specifically target these biological differences in SCLC so normal/non cancer cells are unaffected, leading to decreased side effects and a better quality of life. There is still a lot unknown about SCLC, but these new findings offer a glimmer of hope for patients in the future. Abstract Small cell lung cancer (SCLC) is a high-grade neuroendocrine malignancy with an aggressive behavior and dismal prognosis. 5-year overall survival remains a disappointing 7%. Genomically, SCLCs are homogeneous compared to non-small cell lung cancers and are characterized almost always by functional inactivation of RB1 and TP53 with no actionable mutations. Additionally, SCLCs histologically appear uniform. Thus, SCLCs are currently managed as a single disease with platinum-based chemotherapy remaining the cornerstone of treatment. Recent studies have identified expression of dominant transcriptional signatures which may permit classification of SCLCs into four biologically distinct subtypes, namely, SCLC-A, SCLC-N, SCLC-P, and SCLC-I. These groups are readily detectable by immunohistochemistry and also have potential predictive utility for emerging therapies, including PARPi, immune checkpoint inhibitors, and DLL3 targeted therapies. In contrast with their histology, studies have identified that SCLCs display both inter- and intra-tumoral heterogeneity. Identification of subpopulations of cells with high expression of PLCG2 has been linked with risk of metastasis. SCLCs also display subtype switching under therapy pressure which may contribute furthermore to metastatic ability and chemoresistance. In this review, we summarize the recent developments in the understanding of the biology of SCLCs, and discuss the potential diagnostic, prognostic, and treatment opportunities the four proposed subtypes may present for the future. We also discuss the emerging evidence of tumor heterogeneity and plasticity in SCLCs which have been implicated in metastasis and acquired therapeutic resistance seen in these aggressive tumors.
Collapse
Affiliation(s)
- Anna Keogh
- Department of Histopathology, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 HD53 Dublin, Ireland
- Correspondence:
| | - Stephen Finn
- Department of Histopathology, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 HD53 Dublin, Ireland
| | - Teodora Radonic
- Department of Pathology, Amsterdam University Medical Center, VUMC, University Amsterdam, 1081 HV Amsterdam, The Netherlands;
| |
Collapse
|