1
|
Jiang D, Xiao Y, Yu Z, Deng M, Song Q, Huang J, Su J, Xu C, Hou Y. Unravelling the difference of immune microenvironment characteristics between esophageal basaloid squamous cell carcinoma and conventional esophageal squamous cell carcinoma. Hum Pathol 2025; 155:105716. [PMID: 39793931 DOI: 10.1016/j.humpath.2025.105716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
OBJECTIVES Esophageal basaloid squamous cell carcinoma (basaloid ESCC) is an uncommon variant of esophageal squamous cell carcinoma (ESCC). We characterized the tumor immune microenvironment features of basaloid ESCC, and compared them with conventional ESCC. METHODS AND RESULTS One hundred and four basaloid ESCC patients and 55 conventional ESCC patients were included in Cohort 1. Among 104 basaloid ESCC, 81 were pure basaloid ESCC, and 23 were mixed basaloid ESCC with invasive basaloid ESCC components and ESCC components. In pure basaloid ESCC, there were more immature desmoplastic reaction (P < 0.001), fewer peritumoral tertiary lymphoid structures (TLSs) (P < 0.001), and lower tumor proportional score (TPS) and combined positive score (CPS) (P < 0.001 and P = 0.004) than in conventional ESCC. In mixed basaloid ESCC, the number of mature or intermediate desmoplastic reaction (P < 0.001), the tumor-infiltrating lymphocytes (TILs) score (P = 0.043), the proportion of stromal CD8+ TILs (P = 0.047), the number of intratumoral CD20+ TILs (P < 0.001), the number of peritumoral TLSs (P = 0.001), the number of peritumoral matureTLSs (P = 0.021), and the PD-L1 (22C3) CPS (P = 0.016) were lower in the basaloid ESCC components than in the conventional ESCC components. In addition, the data of 141 ESCC patients with neoadjuvant chemoimmunotherapy (nICT) were collected to compare immunotherapeutic outcomes. Among 141 nICT-treated patients, 115 patients had residual tumor cells remaining, including 101 with conventional ESCC and 11 with basaloid ESCC. In basaloid ESCC, 18.2% patients had less than 10% residual viable tumor in the esophageal wall (effective response), which was lower than 58.6% in conventional ESCC (P = 0.025). CONCLUSIONS Our data indicated that basaloid ESCC had less benefits from immunotherapy than conventional ESCC, and manifested as immune "cold" with immature-type desmoplastic reaction, lower TILs, lower peritumoral TLSs, and lower PD-L1 expression than conventional ESCC.
Collapse
Affiliation(s)
- Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Yuhao Xiao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Zixiang Yu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Minying Deng
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China.
| |
Collapse
|
2
|
Rypens C, Van Berckelaer C, Berditchevski F, van Dam P, Van Laere S. Deciphering the molecular biology of inflammatory breast cancer through molecular characterization of patient samples and preclinical models. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 384:77-112. [PMID: 38637101 DOI: 10.1016/bs.ircmb.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Inflammatory breast cancer is an aggressive subtype of breast cancer with dismal patient prognosis and a unique clinical presentation. In the past two decades, molecular profiling technologies have been used in order to gain insight into the molecular biology of IBC and to search for possible targets for treatment. Although a gene signature that accurately discriminates between IBC and nIBC patient samples and preclinical models was identified, the overall genomic and transcriptomic differences are small and ambiguous, mainly due to the limited sample sizes of the evaluated patient series and the failure to correct for confounding effects of the molecular subtypes. Nevertheless, data collected over the past 20 years by independent research groups increasingly support the existence of several IBC-specific biological characteristics. In this review, these features are classified as established, emerging and conceptual hallmarks based on the level of evidence reported in the literature. In addition, a synoptic model is proposed that integrates all hallmarks and that can explain how cancer cell intrinsic mechanisms (i.e. NF-κB activation, genomic instability, MYC-addiction, TGF-β resistance, adaptive stress response, chromatin remodeling, epithelial-to-mesenchymal transition) can contribute to the establishment of the dynamic immune microenvironment associated with IBC. It stands to reason that future research projects are needed to further refine (parts of) this model and to investigate its clinical translatability.
Collapse
Affiliation(s)
- Charlotte Rypens
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium; CellCarta N V, Wilrijk, Belgium
| | - Christophe Van Berckelaer
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, United Kingdom
| | - Peter van Dam
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium; Multidisciplinary Oncological Centre Antwerp (MOCA), Antwerp University Hospital, Drie Eikenstraat 655, Edegem, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
3
|
De Schepper M, Nguyen HL, Richard F, Rosias L, Lerebours F, Vion R, Clatot F, Berghian A, Maetens M, Leduc S, Isnaldi E, Molinelli C, Lambertini M, Grillo F, Zoppoli G, Dirix L, Punie K, Wildiers H, Smeets A, Nevelsteen I, Neven P, Vincent-Salomon A, Larsimont D, Duhem C, Viens P, Bertucci F, Biganzoli E, Vermeulen P, Floris G, Desmedt C. Treatment Response, Tumor Infiltrating Lymphocytes and Clinical Outcomes in Inflammatory Breast Cancer-Treated with Neoadjuvant Systemic Therapy. CANCER RESEARCH COMMUNICATIONS 2024; 4:186-199. [PMID: 38147006 PMCID: PMC10807408 DOI: 10.1158/2767-9764.crc-23-0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/01/2023] [Accepted: 12/13/2023] [Indexed: 12/27/2023]
Abstract
Inflammatory breast cancer (IBC) is a rare (1%-5%), aggressive form of breast cancer, accounting for approximately 10% of breast cancer mortality. In the localized setting, standard of care is neoadjuvant chemotherapy (NACT) ± anti-HER2 therapy, followed by surgery. Here we investigated associations between clinicopathologic variables, stromal tumor-infiltrating lymphocytes (sTIL), and pathologic complete response (pCR), and the prognostic value of pCR. We included 494 localized patients with IBC treated with NACT from October 1996 to October 2021 in eight European hospitals. Standard clinicopathologic variables were collected and central pathologic review was performed, including sTIL. Associations were assessed using Firth logistic regression models. Cox regressions were used to evaluate the role of pCR and residual cancer burden (RCB) on disease-free survival (DFS), distant recurrence-free survival (DRFS), and overall survival (OS). Distribution according to receptor status was as follows: 26.4% estrogen receptor negative (ER-)/HER2-; 22.0% ER-/HER2+; 37.4% ER+/HER2-, and 14.1% ER+/HER2+. Overall pCR rate was 26.3%, being highest in the HER2+ groups (45.9% for ER-/HER2+ and 42.9% for ER+/HER2+). sTILs were low (median: 5.3%), being highest in the ER-/HER2- group (median: 10%). High tumor grade, ER negativity, HER2 positivity, higher sTILs, and taxane-based NACT were significantly associated with pCR. pCR was associated with improved DFS, DRFS, and OS in multivariable analyses. RCB score in patients not achieving pCR was independently associated with survival. In conclusion, sTILs were low in IBC, but were predictive of pCR. Both pCR and RCB have an independent prognostic role in IBC treated with NACT. SIGNIFICANCE IBC is a rare, but very aggressive type of breast cancer. The prognostic role of pCR after systemic therapy and the predictive value of sTILs for pCR are well established in the general breast cancer population; however, only limited information is available in IBC. We assembled the largest retrospective IBC series so far and demonstrated that sTIL is predictive of pCR. We emphasize that reaching pCR remains of utmost importance in IBC.
Collapse
Affiliation(s)
- Maxim De Schepper
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Ha-Linh Nguyen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - François Richard
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Louise Rosias
- Department of Gynecological and Obstetrics, University Hospitals Leuven, Leuven, Belgium
| | | | - Roman Vion
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France
| | - Florian Clatot
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France
| | - Anca Berghian
- Anatomical Pathology Unit, Department of Biopathology, Centre Henri Becquerel, Rouen, France
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sophia Leduc
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Edoardo Isnaldi
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Chiara Molinelli
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Federica Grillo
- Anatomical Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics, University of Genova, Genoa, Italy
- Department of Internal Medicine and Specialistic Medicine, U.O. Medicina Interna a Indirizzo Oncologico, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gabriele Zoppoli
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Internal Medicine and Specialistic Medicine, U.O. Medicina Interna a Indirizzo Oncologico, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luc Dirix
- Translational Cancer Research Unit, Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, GZA hospitals, Antwerp, Belgium
| | - Kevin Punie
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Ann Smeets
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Ines Nevelsteen
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Anne Vincent-Salomon
- Department of Pathology, Université Paris Sciences Lettres, Institut Curie, Paris, France
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium
| | - Caroline Duhem
- Clinique du sein, Centre Hospitalier du Luxembourg, Luxembourg
| | | | | | - Elia Biganzoli
- Unit of Medical Statistics, Biometry and Epidemiology, Department of Biomedical and Clinical Sciences (DIBIC) “L. Sacco” & DSRC, LITA Vialba campus, University of Milan, Milan, Italy
| | - Peter Vermeulen
- Translational Cancer Research Unit, Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, GZA hospitals, Antwerp, Belgium
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Translational Cell and Tissue Research, Department of Pathology and Imaging, KU Leuven, Belgium
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
4
|
Tseng LM, Huang CC, Tsai YF, Chen JL, Chao TC, Lai JI, Lien PJ, Lin YS, Feng CJ, Chen YJ, Chiu JH, Hsu CY, Liu CY. Correlation of an immune-related 8-gene panel with pathologic response to neoadjuvant chemotherapy in patients with primary breast cancers. Transl Oncol 2023; 38:101782. [PMID: 37713974 PMCID: PMC10506137 DOI: 10.1016/j.tranon.2023.101782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/23/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023] Open
Abstract
Neoadjuvant chemotherapy (NACT)-induced pathologic complete response (pCR) is associated with a favorable prognosis for breast cancer. Prior research links tumor-infiltrating lymphocytes with breast cancer chemotherapy response, suggesting the tumor-immune microenvironment's role. The aim of this study was to evaluate the immune-related genes that exhibit associations with the response to NACT. In this study, we analyzed a total of 37 patients (aged 27-67) who received NACT as the first-line treatment for primary breast cancer, followed by surgery. This group consisted of nine patients (24.3 %) with estrogen receptor (ER)-positive/HER2-negative status, ten patients (27.0 %) with ER-positive/HER2-positive status, five patients (13.5 %) with ER-negative/HER2-positive status, and thirteen patients (35.1 %) with triple-negative breast cancer (TNBC). Among these patients, twelve (32.4 %) achieved a pCR, with eight (66.6 %) having HER2-positive tumors, and the remaining four having TNBC. To identify immune-related genes linked with pCR in subjects with breast cancer prior to NACT, we collected fresh tissues for next-generation sequencing. Patients with pCR had higher expressions of eight genes, KLRK1, IGJ, CD69, CD40LG, MS4A1, CD1C, KLRB1, and CA4, compared to non-pCR patients. The 8-gene signature was associated with good prognosis and linked to better relapse-free survival in patients receiving chemotherapy. The expression of these genes was involved in better drug response, displaying a positive correlation with the infiltration of immune cells. In conclusion, we have identified eight immune-related genes that are associated with a favorable prognosis and positive responses to drugs. This 8-gene signature could potentially provide prognostic insights for breast cancer patients undergoing NACT.
Collapse
Affiliation(s)
- Ling-Ming Tseng
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Experimental Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Cheng Huang
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yi-Fang Tsai
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ji-Lin Chen
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ta-Chung Chao
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jiun-I Lai
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; Center of Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Ju Lien
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Nursing, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Shu Lin
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Jung Feng
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Jen Chen
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jen-Hwey Chiu
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Yi Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Yu Liu
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
5
|
Valenza C, Trapani D, Fusco N, Wang X, Cristofanilli M, Ueno NT, Curigliano G. The immunogram of inflammatory breast cancer. Cancer Treat Rev 2023; 119:102598. [PMID: 37437342 DOI: 10.1016/j.ctrv.2023.102598] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
Inflammatory breast cancer (IBC) is the most aggressive and fatal clinical presentation of breast cancer. Despite the term "inflammatory", based on the clinical presentation, IBC is biologically driven by an immunosuppressive tumor microenvironment (TME). Whether IBC can be switched into an immune-inflamed TME by immune-checkpoint inhibitors (ICIs) is a matter of debate. Presently, measurable biomarkers of IBC-TME have never been synthetized into a comprehensive portray of the immune-milieu (i.e., an immunogram), describing the immune-vulnerability of IBC and potentially predicting the response to ICIs. We propose an immunogram for IBC, based on preclinical and clinical studies, including six parameters: the presence of immune-effector cells, of immune-suppressive cells and of immune checkpoints, the general immune status, the activation of immune-suppressive pathways, the tumor foreignness. The IBC immunogram suggests the existence of a preexisting immune TME that is suppressed by mechanisms of immune-escape but might be restored by ICIs. The combination of chemotherapy and ICIs in patients with IBC is based on a strong biological rationale. However, the design and the development of clinical trials assessing the incorporation of ICIs raise many methodological and practical issues. In parallel with the further comprehension of IBC biology, the prospective validation and integration of biomarkers predictive of response to ICIs are warranted.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Pathology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Xiaoping Wang
- The University of Texas MD Anderson Cancer Center, Department of Breast Medical Oncology, Houston, TX, USA
| | - Massimo Cristofanilli
- Division of Medical Oncology, Internal Medicine Department, Weill Cornell Medicine, New York, NY, USA
| | - Naoto T Ueno
- University of Hawai'i Cancer Center, Honolulu, HI 96813, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
6
|
Hazra A, O’Hara A, Polyak K, Nakhlis F, Harrison BT, Giordano A, Overmoyer B, Lynce F. Copy Number Variation in Inflammatory Breast Cancer. Cells 2023; 12:cells12071086. [PMID: 37048158 PMCID: PMC10093603 DOI: 10.3390/cells12071086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Identification of a unique genomic biomarker in de novo inflammatory breast cancer (IBC) may provide an insight into the biology of this aggressive disease. The goal of our study was to elucidate biomarkers associated with IBC. We examined breast biopsies collected from Dana–Farber Cancer Institute patients with IBC prior to initiating preoperative systemic treatment (30 samples were examined, of which 14 were eligible). Patients without available biopsies (n = 1), with insufficient tumor epithelial cells (n = 10), or insufficient DNA yield (n = 5) were excluded from the analysis. Molecular subtype and tumor grade were abstracted from a medical records’ review. Ten IBC tumors were estrogen-receptor-positive (ER+) and human epidermal growth factor receptor 2 (HER2)-negative (n = 10 out of 14). Sufficient RNA and DNA were simultaneously extracted from 14 biopsy specimens using the Qiagen AllPrep Kit. RNA was amplified using the Sensation kit and profiled using the Affymetrix Human Transcriptome Array 2.0. DNA was profiled for genome-wide copy number variation (CNV) using the Affymetrix OncoScan Array and analyzed using the Nexus Chromosome Analysis Suite. Among the 14 eligible samples, we first confirmed biological concordance and quality control metrics using replicates and gene expression data. Second, we examined CNVs and gene expression change by IBC subtype. We identified significant CNVs in IBC patients after adjusting for multiple comparisons. Next, to assess whether the CNVs were unique to IBC, we compared the IBC CNV data to fresh-frozen non-IBC CNV data from The Cancer Genome Atlas (n = 388). On chromosome 7p11.2, we identified significant CN gain located at position 58,019,983-58,025,423 in 8 ER+ IBC samples compared to 338 non-IBC ER+ samples (region length: 5440 bp gain and 69,039 bp, False Discovery Rate (FDR) p-value = 3.12 × 10−10) and at position 57,950,944–58,025,423 in 3 TN-IBC samples compared to 50 non-IBC TN samples (74,479 base pair, gain, FDR p-value = 4.27 × 10−5; near the EGFR gene). We also observed significant CN loss on chromosome 21, located at position 9,648,315–9,764,385 (p-value = 4.27 × 10−5). Secondarily, differential gene expression in IBC patients with 7p11.2 CN gain compared to SUM149 were explored after FDR correction for multiple testing (p-value = 0.0016), but the results should be interpreted with caution due to the small sample size. Finally, the data presented are hypothesis-generating. Validation of CNVs that contribute to the unique presentation and biological features associated with IBC in larger datasets may lead to the optimization of treatment strategies.
Collapse
Affiliation(s)
- Aditi Hazra
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Inflammatory Breast Cancer Program, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Kornelia Polyak
- Inflammatory Breast Cancer Program, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medical Oncology, Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Faina Nakhlis
- Inflammatory Breast Cancer Program, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Surgery, Division of Breast Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Beth T. Harrison
- Inflammatory Breast Cancer Program, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Antonio Giordano
- Inflammatory Breast Cancer Program, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medical Oncology, Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Beth Overmoyer
- Inflammatory Breast Cancer Program, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medical Oncology, Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Filipa Lynce
- Inflammatory Breast Cancer Program, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medical Oncology, Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Alonso-Miguel D, Fiering S, Arias-Pulido H. Proactive Immunotherapeutic Approaches against Inflammatory Breast Cancer May Improve Patient Outcomes. Cells 2022; 11:2850. [PMID: 36139425 PMCID: PMC9497132 DOI: 10.3390/cells11182850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory breast cancer (IBC) is highly metastatic at the onset of the disease with no IBC-specific treatments, resulting in dismal patient survival. IBC treatment is a clear unmet clinical need. This commentary highlights findings from a recent seminal approach in which pembrolizumab, a checkpoint inhibitor against programmed cell death protein 1 (PD-1), was provided to a triple-negative IBC patient as a neoadjuvant immune therapy combined with anthracycline-taxane-based chemotherapy. We highlight the findings of the case report and offer a perspective on taking a proactive approach to deploy approved immune checkpoint inhibitors. On the basis of our recently published research study, we propose in situ vaccination with direct injection of immunostimulatory agents into the tumor as an option to improve outcomes safely, effectively, and economically for IBC patients.
Collapse
Affiliation(s)
- Daniel Alonso-Miguel
- Department of Animal Medicine and Surgery, Veterinary Medicine School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Steven Fiering
- Department of Microbiology and Immunology, and Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth and Dartmouth Hitchcock Health, Lebanon, NH 03756, USA
| | - Hugo Arias-Pulido
- Department of Microbiology and Immunology, and Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth and Dartmouth Hitchcock Health, Lebanon, NH 03756, USA
| |
Collapse
|