1
|
Ramalingam K, Woody R, Glencer A, Schwartz CJ, Mori H, Wong J, Hirst G, Rosenbluth J, Onishi N, Gibbs J, Hylton N, Borowsky AD, Campbell M, Esserman LJ. Intratumoral Injection of mRNA-2752 and Pembrolizumab for High-Risk Ductal Carcinoma In Situ: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol 2025; 11:288-292. [PMID: 39821301 PMCID: PMC11926637 DOI: 10.1001/jamaoncol.2024.5927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/04/2024] [Indexed: 01/19/2025]
Abstract
Importance Intratumoral immunotherapy that leverages the biological characteristics of high-risk ductal carcinoma in situ (DCIS) may be able to reduce the extent of surgical treatment and provide an alternative approach to improve patient outcomes. Objective To determine if combination intratumoral immunotherapy can activate immune cells to shrink or eliminate high-risk DCIS. Design, Setting, and Participants This phase 1 open-label nonrandomized clinical trial at a single academic center tested the safety and efficacy of intratumoral immunotherapy in patients with high-risk DCIS, defined as at least 2 of the following present: younger than 45 years, tumor size greater than 5 cm, high-grade, palpable mass, hormone receptor (HR)-negative, or ERBB2-positive. Patients were enrolled between June 8, 2021, and December 13, 2022. Intervention Pembrolizumab (anti-programmed cell death protein 1), dose ranging from 2 mg to 8 mg, and mRNA-2752 (a combination of interleukin [IL]-23, IL-36γ, and OX40L mRNAs), dose ranging from 1 mg to 4 mg, delivered intratumorally, with 2 to 4 doses given 2 to 3 weeks apart. Main Outcomes and Measures The primary objective was to evaluate the safety and tolerability of intratumoral injections of pembrolizumab and mRNA-2752. The secondary objectives were to assess radiologic and pathological responses and immunological and histological differences in the posttreatment tumor microenvironment. Results Ten female patients with high-risk DCIS (median [range] age, 46 [35-80] years) were enrolled. The median (range) tumor size was 5.3 (1.0-10.0) cm. Five tumors were HR-negative ERBB2-positive; 2 HR-negative ERBB2-negative; 2 HR-positive ERBB2-negative; and 1 HR-positive ERBB2-positive. Of all treated patients, 8 of 10 responded to treatment, and all 8 patients had ERBB2-positive or HR-negative DCIS. Three patients had complete responses. Three patients with negative posttreatment core biopsy results declined surgery and remained disease-free after 1 to 2 years. Multiplex immunofluorescence staining demonstrated that high baseline levels of tumor-infiltrating lymphocytes and programmed cell death ligand 1-positive cells (immune or tumor) were associated with a better treatment response. All patients experienced up to 1 week of fever, malaise, flulike symptoms, axillary adenopathy, erythema, injection site swelling, and swelling in the breast. One patient had intermittent urticaria for 3 months. The dose was serially reduced from 8 mg to 2 mg for pembrolizumab and 4 mg to 1 mg for mRNA-2752 to improve tolerability. The final recommended combination dose is pembrolizumab, 4 mg, with mRNA-2752, 1 mg. Conclusions and Relevance In this phase 1 nonrandomized clinical trial, the results suggest that intratumoral injections of pembrolizumab and mRNA-2752 are safe and may induce rapid regression of high-risk DCIS with high immune infiltrates. These findings warrant additional investigation, and studies are ongoing. Trial Registration ClinicalTrials.gov Identifier: NCT02872025.
Collapse
MESH Headings
- Humans
- Female
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Middle Aged
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/immunology
- Adult
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Injections, Intralesional
- Aged
- RNA, Messenger/administration & dosage
- RNA, Messenger/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
| | - Rachel Woody
- Department of Surgery, University of California, San Francisco
| | - Alexa Glencer
- Department of Surgery, University of California, San Francisco
| | | | - Hidetoshi Mori
- Department of Pathology and Lab Medicine, University of California, Davis
| | - Jasmine Wong
- Department of Surgery, University of California, San Francisco
| | - Gillian Hirst
- Department of Surgery, University of California, San Francisco
| | | | - Natsuko Onishi
- Dempartment of Radiology, University of California, San Francisco
| | - Jessica Gibbs
- Dempartment of Radiology, University of California, San Francisco
| | - Nola Hylton
- Dempartment of Radiology, University of California, San Francisco
| | | | | | | |
Collapse
|
2
|
Mantooth SM, Abdou Y, Saez-Ibañez AR, Upadhaya S, Zaharoff DA. Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies. Front Immunol 2024; 15:1385484. [PMID: 38803496 PMCID: PMC11128577 DOI: 10.3389/fimmu.2024.1385484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Breast cancer poses one of the largest threats to women's health. Treatment continues to improve for all the subtypes of breast cancer, but some subtypes, such as triple negative breast cancer, still present a significant treatment challenge. Additionally, metastasis and local recurrence are two prevalent problems in breast cancer treatment. A newer type of therapy, immunotherapy, may offer alternatives to traditional treatments for difficult-to-treat subtypes. Immunotherapy engages the host's immune system to eradicate disease, with the potential to induce long-lasting, durable responses. However, systemic immunotherapy is only approved in a limited number of indications, and it benefits only a minority of patients. Furthermore, immune related toxicities following systemic administration of potent immunomodulators limit dosing and, consequently, efficacy. To address these safety considerations and improve treatment efficacy, interest in local delivery at the site of the tumor has increased. Numerous intratumorally delivered immunotherapeutics have been and are being explored clinically and preclinically, including monoclonal antibodies, cellular therapies, viruses, nucleic acids, cytokines, innate immune agonists, and bacteria. This review summarizes the current and past intratumoral immunotherapy clinical landscape in breast cancer as well as current progress that has been made in preclinical studies, with a focus on delivery parameters and considerations.
Collapse
Affiliation(s)
- Siena M. Mantooth
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
| | - Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - David A. Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
3
|
Borowsky A, Glencer A, Ramalingam K, Schindler N, Mori H, Ghule P, Lee K, Nachmanson D, Officer A, Harismendy O, Stein J, Stein G, Evans M, Weaver D, Yau C, Hirst G, Campbell M, Esserman L. Tumor microenvironmental determinants of high-risk DCIS progression. RESEARCH SQUARE 2024:rs.3.rs-4126092. [PMID: 38766192 PMCID: PMC11100907 DOI: 10.21203/rs.3.rs-4126092/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Ductal carcinoma in situ (DCIS) constitutes an array of morphologically recognized intraductal neoplasms in the mammary ductal tree defined by an increased risk for subsequent invasive carcinomas at or near the site of biopsy detection. However, only 15-45% of untreated DCIS cases progress to invasive cancer, so understanding mechanisms that prevent progression is key to avoid overtreatment and provides a basis for alternative therapies and prevention. This study was designed to characterize the tumor microenvironment and molecular profile of high-risk DCIS that grew to a large size but remained as DCIS. All patients had DCIS lesions >5cm in size with at least one additional high-risk feature: young age (<45 years), high nuclear grade, hormone receptor negativity, HER2 positivity, the presence of comedonecrosis, or a palpable mass. The tumor immune microenvironment was characterized using multiplex immunofluorescence to identify immune cells and their spatial relationships within the ducts and stroma. Gene copy number analysis and whole exome DNA sequencing identified the mutational burden and driver mutations, and quantitative whole-transcriptome/gene expression analyses were performed. There was no association between the percent of the DCIS genome characterized by copy number variants (CNAs) and recurrence events (DCIS or invasive). Mutations, especially missense mutations, in the breast cancer driver genes PIK3CA and TP53 were common in this high-risk DCIS cohort (47% of evaluated lesions). Tumor infiltrating lymphocyte (TIL) density was higher in DCIS lesions with TP53 mutations (p=0.0079) compared to wildtype lesions, but not in lesions with PIK3CA mutations (p=0.44). Immune infiltrates were negatively associated with hormone receptor status and positively associated with HER2 expression. High levels of CD3+CD8- T cells were associated with good outcomes with respect to any subsequent recurrence (DCIS or invasive cancer), whereas high levels of CD3+Foxp3+ Treg cells were associated with poor outcomes. Spatial proximity analyses of immune cells and tumor cells demonstrated that close proximity of T cells with tumor cells was associated with good outcomes with respect to any recurrence as well as invasive recurrences. Interestingly, we found that myoepithelial continuity (distance between myoepithelial cells surrounding the involved ducts) was significantly lower in DCIS lesions compared to normal tissue (p=0.0002) or to atypical ductal hyperplasia (p=0.011). Gene set enrichment analysis identified several immune pathways associated with low myoepithelial continuity and a low myoepithelial continuity score was associated with better outcomes, suggesting that gaps in the myoepithelial layer may allow access/interactions between immune infiltrates and tumor cells. Our study demonstrates the immune microenvironment of DCIS, in particular the spatial proximity of tumor cells and T cells, and myoepithelial continuity are important determinants for progression of disease.
Collapse
|
4
|
Abbasi AB, Wu V, Lang JE, Esserman LJ. Precision Oncology in Breast Cancer Surgery. Surg Oncol Clin N Am 2024; 33:293-310. [PMID: 38401911 DOI: 10.1016/j.soc.2023.12.011] [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: 02/26/2024]
Abstract
Outcomes for patients with breast cancer have improved over time due to increased screening and the availability of more effective therapies. It is important to recognize that breast cancer is a heterogeneous disease that requires treatment based on molecular characteristics. Early endpoints such as pathologic complete response correlate with event-free survival, allowing the opportunity to consider de-escalation of certain cancer treatments to avoid overtreatment. This article discusses clinical trials of tailoring treatment (eg, I-SPY2) and screening (eg, WISDOM) to individual patients based on their unique risk features.
Collapse
Affiliation(s)
- Ali Benjamin Abbasi
- Department of Surgery, San Francisco Breast Care Center, University of California, Box 1710, UCSF, San Francisco, CA 94143, USA
| | - Vincent Wu
- Department of Surgery, Cleveland Clinic Breast Services, 9500 Euclid Avenue, A80, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Julie E Lang
- Department of Surgery, Cleveland Clinic Breast Services, 9500 Euclid Avenue, A80, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Laura J Esserman
- Department of Surgery, San Francisco Breast Care Center, University of California, Box 1710, UCSF, San Francisco, CA 94143, USA
| |
Collapse
|
5
|
Gavrielatou N, Fortis E, Spathis A, Anastasiou M, Economopoulou P, Foukas GRP, Lelegiannis IM, Rusakiewicz S, Vathiotis I, Aung TN, Tissot S, Kastrinou A, Kotsantis I, Vagia EM, Panayiotides I, Rimm DL, Coukos G, Homicsko K, Foukas P, Psyrri A. B-cell infiltration is associated with survival outcomes following programmed cell death protein 1 inhibition in head and neck squamous cell carcinoma. Ann Oncol 2024; 35:340-350. [PMID: 38159908 DOI: 10.1016/j.annonc.2023.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1) axis blockade has become the mainstay in the treatment of recurrent and/or metastatic (R/M) head and neck squamous cell cancer (HNSCC). Programmed death-ligand 1 (PD-L1) is the only approved biomarker for patient selection; however, response rate is limited even among high expressors. Our primary objective was to investigate the association of immune cell-related biomarkers in the tumor and tumor microenvironment with PD-1 checkpoint inhibitors' outcomes in patients with R/M HNSCC. PATIENTS AND METHODS NCT03652142 was a prospective study in nivolumab-treated platinum-refractory R/M HNSCC, aiming to evaluate biomarkers of response to treatment. Tumor biopsies and blood samples were collected from 60 patients at baseline, post-treatment, and at progression. Immune cells in the tumor and stromal compartments were quantified by immunofluorescence using a five-protein panel (CD3, CD8, CD20, FoxP3, cytokeratin). Tertiary lymphoid structures (TLSs), PD-L1 expression, and peripheral blood immune cell composition were also evaluated for associations with outcome. Our findings were validated by gene set enrichment analysis (GSEA) messenger RNA in situ expression data from the same patients, for B-cell- and TLS-associated genes. RESULTS High pre-treatment density of stromal B cells was associated with prolonged progression-free survival (PFS) (P = 0.011). This result was validated by GSEA, as stromal enrichment with B-cell-associated genes showed association with response to nivolumab. PD-L1 positivity combined with high B-cell counts in stroma defined a subgroup with significantly longer PFS and overall survival (P = 0.013 and P = 0.0028, respectively). CONCLUSIONS Increased B cells in pre-treatment HNSCC biopsy samples correlate with prolonged benefit from PD-1-based immunotherapy and could further enhance the predictive value of PD-L1 expression.
Collapse
Affiliation(s)
- N Gavrielatou
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece; Department of Pathology, Yale University School of Medicine, New Haven, USA
| | - E Fortis
- Ludwig Institute for Cancer Research, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - A Spathis
- Department of Pathology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - M Anastasiou
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - P Economopoulou
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - G R P Foukas
- Department of Pathology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - I M Lelegiannis
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - S Rusakiewicz
- Ludwig Institute for Cancer Research, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - I Vathiotis
- Department of Pathology, Yale University School of Medicine, New Haven, USA
| | - T N Aung
- Department of Pathology, Yale University School of Medicine, New Haven, USA
| | - S Tissot
- Ludwig Institute for Cancer Research, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - A Kastrinou
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - I Kotsantis
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - E M Vagia
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - I Panayiotides
- Department of Pathology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - D L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, USA
| | - G Coukos
- Ludwig Institute for Cancer Research, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - K Homicsko
- Ludwig Institute for Cancer Research, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - P Foukas
- Department of Pathology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - A Psyrri
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
6
|
Türkmen NB, Yüce H, Şahin Y, Taşlıdere AÇ, Özek DA, Ünüvar S, Çiftçi O. Protective effect of resveratrol against pembrolizumab-induced hepatotoxicity and neurotoxicity in male rats. J Biochem Mol Toxicol 2023; 37:e23263. [PMID: 36419233 DOI: 10.1002/jbt.23263] [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: 05/29/2022] [Revised: 09/23/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022]
Abstract
The present study investigates the effects of resveratrol (RSV) on brain and liver tissues in rats with pembrolizumab (PEMB)-induced toxicity. Obtained for the study were 28 male Sprague-Dawley rats (3-4 months old) which were divided into four groups: Group 1: Control. Group 2: Administered PEMB at 5 mg/kg/day i.p. for a week. Group 3: Administered RSV orally at the dose of 20 mg/kg/day for 30 days by gavage. Group 4: Administered PEMB and RSV at 20 and 5 mg/kg/day RSV, respectively, for 30 days. The results of this study revealed that PEMB leads to a significant increase in thiobarbituric acid reactive substance (TBARS) levels and a significant decrease in glutathione peroxidase (GPx), catalase (CAT), superoxide dismutase (SOD) activities, and glutathione (GSH) levels in the liver and brain tissues. The decreased SOD, CAT, GPx activities, and GSH levels increased significantly following RSV treatment in Group 4. The PEMB treatment showed histopathological alterations associated with strong positive cysteinyl aspartic acid-protease-3 (caspase-3) immunoreactivity, while RSV treatment reduced both the expression of caspase-3 protein and the histopathological changes. RSV administration prevents the biochemical, immunological, and histological alterations induced by PEMB. It can be suggested that the lower caspase-3 immunoreactivity in the PEMB + RSV group than in the PEMB group led to an inhibition of RSV on apoptosis.
Collapse
Affiliation(s)
- Neşe B Türkmen
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Hande Yüce
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Yasemin Şahin
- Department of Medical Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Aslı Ç Taşlıdere
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Dilan A Özek
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Inonu University, Malatya, Turkey.,Department of Pharmacy Services, Kovancilar Vocational School, Firat University, Elazig, Turkey
| | - Songül Ünüvar
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| | - Osman Çiftçi
- Department of Medical Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
7
|
Kaneko K, Nagata H, Yang XY, Ginzel J, Hartman Z, Everitt J, Hughes P, Haystead T, Morse M, Lyerly HK, Osada T. A Non-Invasive Deep Photoablation Technique to Inhibit DCIS Progression and Induce Antitumor Immunity. Cancers (Basel) 2022; 14:cancers14235762. [PMID: 36497243 PMCID: PMC9735847 DOI: 10.3390/cancers14235762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) of the breast is often managed by lumpectomy and radiation or mastectomy, despite its indolent features. Effective non-invasive treatment strategies could reduce the morbidity of DCIS treatment. We have exploited the high heat shock protein 90 (HSP90) activity in premalignant and malignant breast disease to non-invasively detect and selectively ablate tumors using photodynamic therapy (PDT). PDT with the HSP90-targeting photosensitizer, HS201, can not only ablate invasive breast cancers (BCs) while sparing non-tumor tissue, but also induce antitumor immunity. We hypothesized that HS201-PDT would both non-invasively ablate DCIS and prevent progression to invasive BC. We tested in vitro selective uptake and photosensitivity of HS201 in DCIS cell lines compared to the non-selective parental verteporfin, and assessed in vivo antitumor efficacy in mammary fat pad and intraductal implantation models. Selective uptake of HS201 enabled treatment of intraductal lesions while minimizing toxicity to non-tumor tissue. The in vivo activity of HS201-PDT was also tested in female MMTV-neu mice prior to the development of spontaneous invasive BC. Mice aged 5 months were administered HS201, and their mammary glands were exposed to laser light. HS201-PDT delayed the emergence of invasive BC, significantly prolonged disease-free survival (DFS) (p = 0.0328) and tended to improve overall survival compared to the no-treatment control (p = 0.0872). Systemic administration of anti-PD-L1 was combined with HS201-PDT and was tested in a more aggressive spontaneous tumor model, HER2delta16 transgenic mice. A single PDT dose combined with anti-PD-L1 improved DFS compared to the no-treatment control, which was significantly improved with repetitive HS201-PDT given with anti-PD-L1 (p = 0.0319). In conclusion, a non-invasive, skin- and tissue-sparing PDT strategy in combination with anti-PD-L1 antibodies effectively prevented malignant progression of DCIS to invasive BC. This non-invasive treatment strategy of DCIS may be safe and effective, while providing an option to reduce the morbidity of current conventional treatment for patients with DCIS. Clinical testing of HS201 is currently underway.
Collapse
Affiliation(s)
- Kensuke Kaneko
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Rm 433A Box 2606, Durham, NC 27710, USA
| | - Hiroshi Nagata
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Rm 433A Box 2606, Durham, NC 27710, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Rm 433A Box 2606, Durham, NC 27710, USA
| | - Joshua Ginzel
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zachary Hartman
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Rm 433A Box 2606, Durham, NC 27710, USA
| | - Jeffrey Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Philip Hughes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Timothy Haystead
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael Morse
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Rm 433A Box 2606, Durham, NC 27710, USA
| | - Takuya Osada
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Rm 433A Box 2606, Durham, NC 27710, USA
- Correspondence: ; Tel.: +1-919-668-5369
| |
Collapse
|
8
|
O'Keefe TJ, Harismendy O, Wallace AM. Large and diffuse ductal carcinoma in situ: potentially lethal subtypes of "preinvasive" disease. Int J Clin Oncol 2022; 27:121-130. [PMID: 34618239 PMCID: PMC10874643 DOI: 10.1007/s10147-021-02036-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/17/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Trials for DCIS have not explored whether outcomes for patients with large disease burden requiring mastectomy are comparable to those of patients with lumpectomy-amenable disease. We aim to identify whether patients with DCIS larger than 5 cm and diffuse-type DCIS differ in breast cancer mortality (BCM) from patients with disease less than 5 cm. METHODS Patients diagnosed with DCIS in the SEER program were assessed to identify factors prognostic of breast-cancer-specific survival using competing risks regression. RESULTS 44,849 patients met criteria for the cumulative incidence estimate. On competing risks cumulative incidence approximation, the 10-year estimate for BCM for each group was 1.3%, 1.3%, 2.3%, and 5.1%, respectively, and the difference among groups was significant (p = 0.017). On competing risks regression of patients with known covariates, both diffuse-type disease and disease larger than 5 cm (hazard ratio [HR] = 6.2 and 1.7, p = 0.013 and p = 0.042, respectively) were associated with increased risk of BCM. After matching, DCIS > 5 cm and diffuse disease were associated with increased BCM relative to disease < 5 cm (HR = 1.69, p = 0.04). Among patients undergoing mastectomy for disease larger than 5 cm or diffuse disease, the 10-year cumulative incidence for BCM was 0.5% among patients undergoing bilateral mastectomy and 2.4% for patients undergoing unilateral mastectomy. CONCLUSION Patients with large and diffuse DCIS represent uncommon but poorly studied DCIS subgroups with worse prognoses than patients with disease smaller than 5 cm. Further studies are needed to elucidate the appropriate treatment for these patients.
Collapse
Affiliation(s)
- Thomas J O'Keefe
- Division of Breast Surgery and The Comprehensive Breast Health Center, University of California San Diego, 0819, 3855 Health Sciences Dr, La Jolla, CA, 92037, USA.
| | - Olivier Harismendy
- Division of Biomedical Informatics, Department of Medicine, Moores Cancer Center, University of California San Diego, 3855 Health Sciences Dr, La Jolla, CA, 92037, USA
| | - Anne M Wallace
- Division of Breast Surgery and The Comprehensive Breast Health Center, University of California San Diego, 0819, 3855 Health Sciences Dr, La Jolla, CA, 92037, USA
| |
Collapse
|