1
|
Pei YY. Camrelizumab vs Placebo in Patients With Triple-Negative Breast Cancer. JAMA 2025; 333:1835. [PMID: 40266585 DOI: 10.1001/jama.2025.1291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Affiliation(s)
- Yuan-Yuan Pei
- Clinical Data Center, Guangzhou Medical University Affiliated Women and Children's Medical Center, Guangzhou, China
| |
Collapse
|
2
|
Lee SY, Yoo TK, Lee SB, Kim J, Chung IY, Ko BS, Kim HJ, Lee JW, Son BH. Prognostic value of residual cancer burden after neoadjuvant chemotherapy in breast cancer: a comprehensive subtype-specific analysis. Sci Rep 2025; 15:13977. [PMID: 40263332 PMCID: PMC12015579 DOI: 10.1038/s41598-025-98176-9] [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: 10/21/2024] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
This study evaluated the prognostic impact of residual cancer burden (RCB) on breast cancer subtypes following neoadjuvant chemotherapy (NAC). We retrospectively examined 2,416 breast cancer patients treated with NAC and surgery at Asan Medical Center (2015-2020). Baseline characteristics, clinicopathological parameters, recurrence, and survival outcomes were analyzed using Kaplan-Meier and Cox regression methods to assess RCB's prognostic significance across subtypes. Pathologic complete response (pCR) was achieved in 25.6% (619) of patients. RCB2 was the most common (44.0%, 1,063), followed by RCB3 (19.6%, 474) and RCB1 (10.8%, 260). Among HR-/HER2 + patients, 67% had RCB0/1, while 87% of HR+/HER2- patients had RCB2/3. Higher RCB was significantly associated with worse overall survival (OS) and disease-free survival (DFS) across all subtypes. Subtype-specific analysis revealed that HR-/HER2 + patients with RCB3 and HR-/HER2- patients with RCB2/3 had significantly worse OS and DFS. Multivariate analysis revealed that RCB2/3 (vs. RCB0), total mastectomy (vs. breast-conserving surgery), axillary lymph node dissection (ALND), lymphovascular invasion (LVI), high Ki-67 index (≥ 20), HR negativity, and HER2 negativity were linked to higher risks of recurrence and death (p < 0.05). Factors associated with higher RCB included ALND, LVI, higher Ki-67, and HR+/HER2- subtype. RCB classification is a strong prognostic indicator across all subtypes. Patients with RCB2/3 in the HR-/HER2- and RCB3 in the HR-/HER2 + subtypes had particularly poor outcomes, suggesting benefits from additional treatments beyond standard care.
Collapse
Affiliation(s)
- Soo-Young Lee
- Department of General Surgery, Inha University College of Medicine, Inha University Hospital, Incheon, Korea
| | - Tae-Kyung Yoo
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Sae Byul Lee
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Jisun Kim
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Il Yong Chung
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Beom Seok Ko
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Jong Won Lee
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea
| | - Byung Ho Son
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-gil, Songpa-Gu, Seoul, 05505, Korea.
| |
Collapse
|
3
|
Dennis S, Tsukioki T, Kocherginsky M, Qi AK, DeHorn S, Gurley M, Wrubel E, Luo Y, Khan SA. Neoadjuvant Chemotherapy Response and Genetic Susceptibility in Recently Parous Women with Breast Cancer: A Retrospective Analysis. Ann Surg Oncol 2025:10.1245/s10434-025-17204-0. [PMID: 40205151 DOI: 10.1245/s10434-025-17204-0] [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: 12/05/2024] [Accepted: 03/03/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Women with recent parity are at increased short-term breast cancer (BC) risk and face a worse prognosis. The effect of parity on response to neoadjuvant chemotherapy (NAC) is unstudied, and the influence of inherited susceptibility on parity-related short-term risk remains unclear. METHODS A retrospective case-cohort study analyzed women age 50 years or younger with non-metastatic BC diagnosed between 2010 and 2020 who underwent genetic testing and were treated at Northwestern Medicine. Associations between NAC response and recency of parity were evaluated using multivariate logistic regression, stratified by tumor biologic subtypes. Relationships between germline mutations, recency of parity, and BC were explored via multi-state modeling and linear regression. RESULTS Among 1080 eligible women, 231 received NAC. Treatment response was poorer in parous women with triple-negative tumors than in nullipara women regardless of the recency of parity (P < 0.03). Among 122 women (11.3%) with detectable pathogenic mutations, adjusted analyses with both modeling approaches showed no indications that BRCA1/2 carriers had a greater hazard of a BC diagnosis in the decade after recent parity than nulliparous mutation carriers. For BRCA2 and PALB2 carriers, BC diagnosis occurred less frequently in the postpartum intervals. CONCLUSION This study showed a poor response to NAC in parous triple-negative BC (TNBC) patients than in nullipara patients. The effects of immunotherapy-based regimens deserve evaluation in the context of parity. Postpartum BC occurrence is not increased in BRCA1/2 carriers. The effects of rarer susceptibility genes may differ. These important effects of parity on BC in young women and those at genetic risk warrant larger prospective studies.
Collapse
Affiliation(s)
- Saya Dennis
- Division of Biostatistics and Informatics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Takahiro Tsukioki
- Department of Breast and Thyroid Surgery, Okayama University Hospital, Okayama, Japan
| | - Masha Kocherginsky
- Division of Biostatistics and Informatics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Andrea Keya Qi
- Division of Biostatistics and Informatics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sarah DeHorn
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael Gurley
- Clinical and Translational Sciences Institute, Northwestern University, Evanston, IL, USA
| | - Erica Wrubel
- SHMG Comprehensive Breast Clinic, Grand Rapids, MI, USA
| | - Yuan Luo
- Division of Biostatistics and Informatics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Seema Ahsan Khan
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
4
|
Uematsu M, Nakajima H, Miyake H, Wakabayashi M, Funasaka C, Kondoh C, Harano K, Matsubara N, Hosono A, Naito Y, Sakamoto N, Kojima M, Onishi T, Ishii G, Mukohara T. Digitally quantified area of residual tumor after neoadjuvant chemotherapy in HER2-positive breast cancer. Breast Cancer 2025:10.1007/s12282-025-01694-7. [PMID: 40172786 DOI: 10.1007/s12282-025-01694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The area of residual tumor (ART) is a quantitative method for assessing tumors after neoadjuvant chemotherapy (NAC). This study evaluated whether ART can identify a favorable prognosis group in patients with HER2-positive surgically resected breast cancer and residual tumors post-NAC. METHODS We retrospectively reviewed patients with HER2-positive who underwent surgery after NAC, including trastuzumab, from 2005 to 2022 at our institution. ART was assessed at the maximum cut surface of the residual primary tumor using digital pathology images. Receiver operating characteristic curve analysis determined ART-Low and ART-High cutoffs, excluding ART-0 (0 mm2) patients. RESULTS Of the 219 patients, 82 had ART greater than 0 mm2. The median follow-up was 90.2 months. The number of patients in the ART-0, ART-Low (0 < ART ≤ 4.0 mm2), and ART-High (> 4.0 mm2) groups were 137, 39, and 43, respectively. The ART-Low group showed significantly shorter event-free survival compared to the ART-0 group (HR 3.50, 95% CI 1.52-8.06), and the ART-High group also tended toward poorer prognosis (HR 2.31, 95% CI 0.89-5.97). However, there was no significant difference in prognosis between the ART-Low and ART-High groups. CONCLUSIONS The current study suggests that even minimal residual tumor cells in the primary site can significantly impact on prognosis in HER2-positive early breast cancer.
Collapse
Affiliation(s)
- Mao Uematsu
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
| | - Hiromichi Nakajima
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
- Department of General Internal Medicine, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
| | - Hirohiko Miyake
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Masashi Wakabayashi
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Chuo-Ku, Japan
| | - Chikako Funasaka
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Chihiro Kondoh
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Kenichi Harano
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Ako Hosono
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Pediatric Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Yoichi Naito
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of General Internal Medicine, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Exploratory Oncology Research and Clinical Trial Center, Division of Pathology, Chuo-Ku, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Tatsuya Onishi
- Department of Breast Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Genichiro Ishii
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
| |
Collapse
|
5
|
Budzyń M, Kubicka A, Kaja E, Kycler W, Załuska-Kusz J, Brzeziński JJ, Sperling M, Bukowska A, Grupińska J. Significance of the monocyte CCR2-CCL2 axis in triple-negative breast cancer. Arch Med Res 2025; 56:103205. [PMID: 40073677 DOI: 10.1016/j.arcmed.2025.103205] [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: 09/10/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The monocyte CCR2-CCL2 axis appears to play a crucial role in the generation of tumor-associated macrophages (TAMs), which subsequently promotes tumor metastasis and resistance to therapy. AIMS Our study assessed the monocyte CCR2-CCL2 axis in triple-negative breast cancer (TNBC) and its ability to predict tumor response to neoadjuvant chemotherapy (NAC). METHODS The study included 42 female patients diagnosed with TNBC and eligible for NAC. Response to neoadjuvant chemotherapy was based on pathological complete response (pCR). Surface expression of CCR2 on monocytes was evaluated by flow cytometry. Circulating CCL2 was measured by Luminex X-Map technology. RESULTS Increased monocyte CCR2 expression and higher circulating CCL2 levels were observed in the patients with TNBC. After dividing the patients according to their response to NAC, a significant difference in CCL2 levels was found only between patients who achieved pCR and those who did not. ROC curves showed that the optimum diagnostic cut-off value of CCL2 ≤89.61 pg/mL better discriminated patients with TNBC who achieved pCR better than the Ki-67 index. Univariate analysis demonstrated that circulating. CCL2 ≤89.61 pg/mL was significantly associated with pCR. However, this correlation lost statistical significance in the multivariate model. CONCLUSIONS Our study demonstrated the activation of the monocyte CCR2-CCL2 axis in TNBC for the first time. This activation occurs mainly in patients who do not respond to NAC. Circulating CCL2 levels ≤89.61 pg/mL were found to predict, to some extent, the achievement of pCR in patients with TNBC receiving NAC.
Collapse
Affiliation(s)
- Magdalena Budzyń
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | - Agata Kubicka
- Departament of Cancer Pathology, Greater Poland Cancer Centre, Poznań, Poland; Departament of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, Poznań, Poland
| | - Elżbieta Kaja
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | - Witold Kycler
- Department of Oncological Surgery of Gastrointestinal Diseases, Greater Poland Cancer Centre, Poznań, Poland
| | - Joanna Załuska-Kusz
- Department of Clinical Oncology and Immuno-oncology, Greater Poland Cancer Center, Poznań, Poland
| | - Jacek J Brzeziński
- Department of Oncological Surgery of Gastrointestinal Diseases, Greater Poland Cancer Centre, Poznań, Poland
| | - Marcelina Sperling
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | - Alicja Bukowska
- Medical Analysis Laboratory, Regional Blood Center, Poznań, Poland
| | - Joanna Grupińska
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland; Hospital Pharmacy, Greater Poland Cancer Center, Poznań, Poland
| |
Collapse
|
6
|
Dennis S, Tsukioki T, Kocherginsky M, Qi AK, DeHorn S, Gurley M, Wrubel E, Luo Y, Khan SA. Neoadjuvant chemotherapy response and genetic susceptibility in recently parous women with breast cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.13.25322229. [PMID: 39990588 PMCID: PMC11844593 DOI: 10.1101/2025.02.13.25322229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Introduction Women with recent parity are at increased short-term breast cancer (BC) risk and face a worse prognosis. The effect of parity on response to neoadjuvant chemotherapy (NAC) is unstudied, and the influence of inherited susceptibility on parity-related short-term risk remains unclear. Methods We conducted a retrospective case-cohort study among women aged ≤50 with non-metastatic BC diagnosed between 2010 and 2020 who underwent genetic testing and were treated at Northwestern Medicine. Associations between NAC response and recency of parity were evaluated using multivariate logistic regression, stratified by tumor biologic subtypes. Relationships between germline mutations, recency of parity, and BC were explored via multi-state modeling and linear regression. Results Among 1,080 eligible women, 231 received NAC. Treatment response was poorer in parous women with triple negative tumors compared to nullipara, regardless of the recency of parity ( P <0.03). Among 122 women (11.3%) with detectable pathogenic mutations, adjusted analyses with both modeling approaches revealed no indications that BRCA1/2 carriers had an increased hazard of BC diagnosis in the decade following recent parity, compared to nulliparous mutation carriers. For BRCA2 and PALB2 carriers, breast cancer diagnosis occurred less frequently in the post-partum intervals. Conclusion We observed a poor response to NAC in parous TNBC patients compared to nullipara; effects of immunotherapy-based regimens deserve evaluation in the context of parity. Post-partum BC occurrence is not increased in BRCA1/2 carriers; effects of rarer susceptibility genes may differ. These important effects of parity on BC in young women and those at genetic risk warrant larger prospective studies.
Collapse
|
7
|
Zawati I, Troujette Y, Adouni O, Manai M, Nouira M, Mekki K, Manai M, Rahal K, Gamoudi A. Can residual proliferative cancer burden predict long-term outcomes following neoadjuvant chemotherapy in breast cancer? Pathology 2025:S0031-3025(25)00063-7. [PMID: 40121151 DOI: 10.1016/j.pathol.2024.11.014] [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: 09/20/2023] [Revised: 11/04/2024] [Accepted: 11/26/2024] [Indexed: 03/25/2025]
Abstract
Residual proliferative cancer burden (RPCB) has been suggested as a strong predictor model of long-term outcomes in breast cancer undergoing neoadjuvant chemotherapy (NACT). In our study, we aimed to compare the prognostic value of multiple post-NACT classifications for assessing residual disease. Archival surgical specimens of 97 patients with primary breast cancer who underwent NACT were evaluated for residual cancer burden (RCB). The post-operative Ki-67 proliferation index was quantified using immunohistochemistry on post-treatment surgical excision specimens with residual disease. Then, we calculated the RPCB scores by combining the anatomical RCB index with the biological post-therapeutic Ki-67 using the Cox proportional hazard model for each parameter. Using the Kaplan-Meier method, RCBIII showed an unfavourable prognosis with worse relapse-free survival (RFS) (estimated 5-year RFS rate of 38%) than RCBI, which displayed a similarly good prognosis as pathological complete response (equal to RCB0) (estimated 5-year RFS rates of 80% and 100%, respectively) (p=0.012). The RCBII showed an intermediate prognosis (estimated 5-year RFS rate of 79%). A higher post-NACT Ki-67 (greater than cut-off 20%) had a negative impact on the overall survival and RFS (p<0.0001 for both) using the Kaplan-Meier method. In multivariate analysis, the histological residual tumour size, number of affected lymph nodes, and RCB index remained independent prognostic factors for RFS. In addition, RPCBIII showed the worst prognosis (with an estimated 5-year RFS rate of 38%) compared to RPCBI (estimated 5-year RFS rate of 83%) (p=0.039) by the Kaplan-Meier method. The area under the curve of the RCB index was 0.82 compared to 0.62 for the RPCB model in terms of RFS prediction. Our study highlighted the potential stratification of RCBII cases based on the RPCB classification. Further studies with larger cohorts will be needed to validate whether the RCPB adds value to residual disease assessment.
Collapse
Affiliation(s)
- Imen Zawati
- Department of Immuno-Histo-Cytology, Salah Azaiez Institute, Tunis, Tunisia; Department of Biology, Laboratory of Biochemistry and Molecular Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia.
| | - Yousra Troujette
- Department of Immuno-Histo-Cytology, Salah Azaiez Institute, Tunis, Tunisia
| | - Olfa Adouni
- Department of Immuno-Histo-Cytology, Salah Azaiez Institute, Tunis, Tunisia; Department of Biology, Laboratory of Biochemistry and Molecular Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Maroua Manai
- Department of Immuno-Histo-Cytology, Salah Azaiez Institute, Tunis, Tunisia; Laboratory of Transmission, Control and Immunobiology of Infections - LR16IPT02, Pasteur Institute of Tunis, University of Tunis, Tunis, Tunisia
| | - Meriem Nouira
- Department of Epidemiology and Community Medicine, Charles Nicoles Hospital, Tunis, Tunisia
| | | | - Mohamed Manai
- Department of Biology, Laboratory of Biochemistry and Molecular Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Khaled Rahal
- Department of Surgical Oncology, Salah Azaiez Institute, Tunis, Tunisia
| | - Amor Gamoudi
- Department of Immuno-Histo-Cytology, Salah Azaiez Institute, Tunis, Tunisia
| |
Collapse
|
8
|
Jia Z, Xing H, Wang J, Wang X, Wang X, Liu C, He J, Wu S, Miao J, Liu H, Liu Y. Prognostic factors of patients with human epidermal growth factor receptor 2-positive breast cancer following neoadjuvant therapy: Development and validation of a predictive nomogram. Pathol Res Pract 2024; 261:155504. [PMID: 39116570 DOI: 10.1016/j.prp.2024.155504] [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: 09/09/2023] [Revised: 05/21/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Human epidermal growth factor receptor 2 (HER2)-positive breast cancer exhibits an aggressive phenotype and poor prognosis. The application of neoadjuvant therapy (NAT) in patients with breast cancer can significantly reduce the risks of disease recurrence and improve survival. By integrating different clinicopathological factors, nomograms are valuable tools for prognosis prediction. This study aimed to assess the prognostic value of clinicopathological factors in patients with HER2-positive breast cancer and construct a nomogram for outcome prediction. METHODS We retrospectively analyzed the clinicopathological data from 374 patients with breast cancer admitted to the Fourth Hospital of Hebei Medical University between January 2009 and December 2017, who were diagnosed with invasive breast cancer through preoperative core needle biopsy pathology, underwent surgical resection after NAT, and were HER2-positive. Patients were randomly divided into a training and validation set at a ratio of 7:3. Univariate and multivariate survival analyses were performed using Kaplan-Meier and Cox proportional hazards regression models. Results of the multivariate analysis were used to create nomograms predicting 3-, 5-, and 8-year overall survival (OS) rates. Calibration curves were plotted to test concordance between the predicted and actual risks. Harrell C-index and time-dependent receiver operating characteristic (ROC) curves were used to evaluate the discriminability of the nomogram prediction model. RESULTS All included patients were women, with a mean age of 50 ± 10.4 years (range: 26-72 years). In the training set, both univariate and multivariate analyses identified residual cancer burden (RCB) class, tumor-infiltrating lymphocytes(TILs), and clinical stage as independent prognostic factors for OS, and these factors were combined to construct a nomogram. The calibration curves demonstrated good concordance between the predicted and actual risks, and the C-index of the nomogram was 0.882 (95 % CI 0.863-0.901). The 3-, 5-, and 8-year areas under the ROC curve (AUCs) were 0.909, 0.893, and 0.918, respectively, indicating good accuracy of the nomogram. The calibration curves also demonstrated good concordance in the validation set, with a C-index of 0.850 (95 % CI 0.804-0.896) and 3-, 5-, and 8-year AUCs of 0.909, 0.815, and 0.834, respectively, which also indicated good accuracy. CONCLUSION The nomogram prediction model accurately predicted the prognostic status of post-NAT patients with breast cancer and was more accurate than clinical stage and RCB class. Therefore, it can serve as a reliable guide for selecting clinical treatment measures for breast cancer.
Collapse
Affiliation(s)
- Zhanli Jia
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Hui Xing
- Department of Pathology, Cangzhou Integrated Traditional Chinese and Western Medicine Hospital, Cangzhou, Hebei 061000, China
| | - Jian Wang
- Department of Urology Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xinran Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xu Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Chang Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jiankun He
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Si Wu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jiaxian Miao
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Hongbo Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China.
| |
Collapse
|
9
|
Thomas N, Garaud S, Langouo M, Sofronii D, Boisson A, De Wind A, Duwel V, Craciun L, Larsimont D, Awada A, Willard-Gallo K. Tumor-Infiltrating Lymphocyte Scoring in Neoadjuvant-Treated Breast Cancer. Cancers (Basel) 2024; 16:2895. [PMID: 39199667 PMCID: PMC11352458 DOI: 10.3390/cancers16162895] [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: 07/10/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Neoadjuvant chemotherapy (NAC) is now the standard of care for patients with locally advanced breast cancer (BC). TIL scoring is prognostic and adds predictive value to the residual cancer burden evaluation after NAC. However, NAC induces changes in the tumor, and the reliability of TIL scoring in post-NAC samples has not yet been studied. H&E- and dual CD3/CD20 chromogenic IHC-stained tissues were scored for stromal and intra-tumoral TIL by two experienced pathologists on pre- and post-treatment BC tissues. Digital TIL scoring was performed using the HALO® image analysis software (version 2.2). In patients with residual disease, we show a good inter-pathologist correlation for stromal TIL on H&E-stained tissues (CCC value 0.73). A good correlation for scoring with both staining methods (CCC 0.81) and the digital TIL scoring (CCC 0.77) was also observed. Overall concordance for TIL scoring in patients with a complete response was however poor. This study reveals there is good reliability for TIL scoring in patients with detectable residual tumors after NAC treatment, which is comparable to the scoring of untreated breast cancer patients. Based on the good consistency observed with digital TIL scoring, the development of a validated algorithm in the future might be advantageous.
Collapse
Affiliation(s)
- Noémie Thomas
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Soizic Garaud
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Mireille Langouo
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Doïna Sofronii
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Anaïs Boisson
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Alexandre De Wind
- Anantomical Pathology Department, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Valérie Duwel
- Anatomical Pathology Department, AZ Klina, 2930 Brasschaat, Belgium;
| | - Ligia Craciun
- Anantomical Pathology Department, Institut Jules Bordet, 1070 Brussels, Belgium
- Tumor Bank, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Dennis Larsimont
- Anantomical Pathology Department, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Ahmad Awada
- Medical Oncology, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| |
Collapse
|
10
|
Quirke NP, Cullinane C, Turk MA, Shafique N, Evoy D, Geraghty J, McCartan D, Quinn C, Walshe JM, McDermott E, Rutherford C, Prichard RS. Invasive lobular carcinoma of the breast; clinicopathologic profile and response to neoadjuvant chemotherapy over a 15-year period. Breast 2024; 76:103739. [PMID: 38754140 PMCID: PMC11126855 DOI: 10.1016/j.breast.2024.103739] [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: 09/11/2023] [Revised: 04/03/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024] Open
Abstract
INTRODUCTION Invasive lobular carcinoma (ILC) accounts for 5-15% of invasive breast cancers. Typical ILC is oestrogen receptor (ER) positive and human epidermal growth factor receptor 2 (HER2) negative. Atypical biomarker profiles (ER- and HER2+, ER+ and HER2+ or triple negative) appear to differ from typical ILCs. This study compared subtypes of ILC in terms of clinical and pathological parameters, and response to neoadjuvant chemotherapy (NACT) according to biomarker profile. METHODS All patients with ILC treated in a single centre from January 2005 to December 2020 were identified from a prospectively maintained database. Clinicopathologic and outcome data was collected and analysed according to tumour biomarker profile. RESULTS A total of 582 patients with ILC were treated. Typical ILC was observed in 89.2% (n = 519) and atypical in 10.8% (n = 63). Atypical ILCs were of a higher grade (35% grade 3 vs 9.6% grade 3, p < 0.001). A larger proportion of atypical ILC received NACT (31.7% vs 6.9% p < 0.001). Atypical ILCs showed a greater response to NACT (mean RCB (Residual Cancer Burden Score) 2.46 vs mean RCB 3.41, p = 0.0365), and higher pathological complete response rates (15% vs 0% p = 0.017). Despite this, overall 5-year disease-free survival (DFS) was higher in patients with typical ILC (91% vs 83%, p = 0.001). CONCLUSIONS Atypical ILCs have distinct characteristics. They are more frequently of a higher grade and demonstrate a superior response to NACT. Despite the latter, atypical ILCs have a worse 5-year DFS which should be taken into consideration in terms of prognostication and may assist patient selection for NACT.
Collapse
Affiliation(s)
- N P Quirke
- UCD School of Medicine, University College Dublin, D04 V1W8, Dublin, Ireland.
| | - C Cullinane
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - M A Turk
- UCD School of Medicine, University College Dublin, D04 V1W8, Dublin, Ireland
| | - N Shafique
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - D Evoy
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - J Geraghty
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - D McCartan
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - C Quinn
- Department of Pathology, St. Vincent's University Hospital, Dublin, Ireland
| | - J M Walshe
- Department of Oncology, St. Vincent's University Hospital, Dublin, Ireland
| | - E McDermott
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - C Rutherford
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - R S Prichard
- Department of Breast and Endocrine Surgery, St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
11
|
Carroll JF, Hoskin TL, Leon-Ferre RA, Boughey JC. Impact of Presenting Stage on Overall Survival in Patients Treated with Neoadjuvant Chemotherapy for Triple Negative Breast Cancer. Ann Surg Oncol 2024; 31:5132-5140. [PMID: 38872043 DOI: 10.1245/s10434-024-15583-4] [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: 03/22/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE For operable triple-negative breast cancer (TNBC) treated with neoadjuvant chemotherapy (NAC), clinical prognostication and postoperative decision-making relies exclusively on whether a pathologic complete response (pCR) is achieved or not. We evaluated whether extent of disease at presentation further influenced overall survival (OS) among patients with pCR or with residual disease (RD) following NAC. METHODS Patients with stage I-III TNBC who underwent NAC were identified from the National Cancer Database from 2010 to 2019. Overall survival was assessed by disease extent using the Kaplan-Meier method and Cox proportional hazards regression for univariate and multivariable analysis. RESULTS A total of 35,598 patients met inclusion criteria, and 11,967 achieved pCR. Ten-year OS was 88.5% and varied by cT and cN category at presentation. Best 10-year OS was seen in patients with cT1-2, cN0 (90.9%) and was worst in those with cT3-4, cN2-3 disease (72.0%). A total of 23,631 patients had RD. Ten-year OS was 60.1% and varied by cT and cN category at presentation. Best 10-year OS was seen in patients with cT1-2, cN0 (73.0%) and was worst in those with cT3-4, cN2-3 disease (36.3%). Notably, OS was significantly poorer for patients with cT3-4, cN2-3 disease at diagnosis and pCR versus those with cT1-2 cN0 and RD (aHR 1.30, 95% confidence interval 1.03-1.63, p = 0.03). CONCLUSIONS Among patients with TNBC, extent of disease at presentation was prognostic for OS independently of response to NAC. Patients with advanced stage at presentation had poorer OS even in the context of pCR. Further investigation is needed to evaluate whether additional adjuvant therapy strategies should be considered for these patients.
Collapse
Affiliation(s)
- Jennifer F Carroll
- Division of Breast and Melanoma Surgical Oncology, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Tanya L Hoskin
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Roberto A Leon-Ferre
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Judy C Boughey
- Division of Breast and Melanoma Surgical Oncology, Department of Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
12
|
Cabioglu N, Onder S, Karatay H, Bayram A, Oner G, Tukenmez M, Muslumanoglu M, Igci A, Dinccag A, Ozmen V, Aydiner A, Saip P, Yavuz E. New Emerging Chemokine Receptors: CCR5 or CXCR5 on Tumor Is Associated with Poor Response to Chemotherapy and Poor Prognosis in Locally Advanced Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:2388. [PMID: 39001456 PMCID: PMC11240792 DOI: 10.3390/cancers16132388] [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: 05/29/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND We aim to investigate any possible associations between chemokine receptor expression and responses to neoadjuvant chemotherapy (NAC) along with outcomes in patients with triple-negative breast cancer (TNBC) with locally advanced disease. METHOD Expressions of chemokine receptors were examined immunohistochemically after staining archival tissue of surgical specimens (n = 63) using specific antibodies for CCR5, CCR7, CXCR4, and CXCR5. RESULTS Patients with high CCR5, CCR7, CXCR4, and CXCR5 expression on tumors and high CXCR4 expression on tumor-infiltrating lymphocytes (TILs) were less likely to have a pathological complete response (pCR) or Class 0-I RCB-Index compared to others. Patients with residual lymph node metastases (ypN-positive), high CCR5TM(tumor), and high CXCR4TM expressions had an increased hazard ratio (HR) compared to others (DFS: HR = 2.655 [1.029-6.852]; DSS: HR = 2.763 [1.008-7.574]), (DFS: HR = 2.036 [0.805-5.148]; DSS: HR = 2.689 [1.020-7.090]), and (DFS: HR = 2.908 [1.080-7.829]; DSS: HR = 2.132 (0.778-5.846)), respectively. However, patients without CXCR5TIL expression had an increased HR compared to those with CXCR5TIL (DFS: 2.838 [1.266-6.362]; DSS: 4.211 [1.770-10.016]). CONCLUSIONS High expression of CXCR4TM and CCR5TM was found to be associated with poor prognosis, and CXCR5TM was associated with poor chemotherapy response in the present cohort with locally advanced TNBC. Our results suggest that patients with TNBC could benefit from a chemokine receptor inhibitor therapy containing neoadjuvant chemotherapy protocols.
Collapse
Affiliation(s)
- Neslihan Cabioglu
- Department of General Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (G.O.); (M.T.); (M.M.); (A.I.); (A.D.); (V.O.)
| | - Semen Onder
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (S.O.); (H.K.); (A.B.); (E.Y.)
| | - Hüseyin Karatay
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (S.O.); (H.K.); (A.B.); (E.Y.)
| | - Aysel Bayram
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (S.O.); (H.K.); (A.B.); (E.Y.)
| | - Gizem Oner
- Department of General Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (G.O.); (M.T.); (M.M.); (A.I.); (A.D.); (V.O.)
| | - Mustafa Tukenmez
- Department of General Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (G.O.); (M.T.); (M.M.); (A.I.); (A.D.); (V.O.)
| | - Mahmut Muslumanoglu
- Department of General Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (G.O.); (M.T.); (M.M.); (A.I.); (A.D.); (V.O.)
| | - Abdullah Igci
- Department of General Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (G.O.); (M.T.); (M.M.); (A.I.); (A.D.); (V.O.)
| | - Ahmet Dinccag
- Department of General Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (G.O.); (M.T.); (M.M.); (A.I.); (A.D.); (V.O.)
| | - Vahit Ozmen
- Department of General Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (G.O.); (M.T.); (M.M.); (A.I.); (A.D.); (V.O.)
| | - Adnan Aydiner
- Department of Medical Oncology, Institute of Oncology, Istanbul University, Istanbul 34452, Turkey; (A.A.); (P.S.)
| | - Pınar Saip
- Department of Medical Oncology, Institute of Oncology, Istanbul University, Istanbul 34452, Turkey; (A.A.); (P.S.)
| | - Ekrem Yavuz
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey; (S.O.); (H.K.); (A.B.); (E.Y.)
| |
Collapse
|
13
|
Bossuyt V, Provenzano E, Symmans WF, Webster F, Allison KH, Dang C, Gobbi H, Kulka J, Lakhani SR, Moriya T, Quinn CM, Sapino A, Schnitt S, Sibbering DM, Slodkowska E, Yang W, Tan PH, Ellis I. A dedicated structured data set for reporting of invasive carcinoma of the breast in the setting of neoadjuvant therapy: recommendations from the International Collaboration on Cancer Reporting (ICCR). Histopathology 2024; 84:1111-1129. [PMID: 38443320 DOI: 10.1111/his.15165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/11/2024] [Indexed: 03/07/2024]
Abstract
AIMS The International Collaboration on Cancer Reporting (ICCR), a global alliance of major (inter-)national pathology and cancer organisations, is an initiative aimed at providing a unified international approach to reporting cancer. ICCR recently published new data sets for the reporting of invasive breast carcinoma, surgically removed lymph nodes for breast tumours and ductal carcinoma in situ, variants of lobular carcinoma in situ and low-grade lesions. The data set in this paper addresses the neoadjuvant setting. The aim is to promote high-quality, standardised reporting of tumour response and residual disease after neoadjuvant treatment that can be used for subsequent management decisions for each patient. METHODS The ICCR convened expert panels of breast pathologists with a representative surgeon and oncologist to critically review and discuss current evidence. Feedback from the international public consultation was critical in the development of this data set. RESULTS The expert panel concluded that a dedicated data set was required for reporting of breast specimens post-neoadjuvant therapy with inclusion of data elements specific to the neoadjuvant setting as core or non-core elements. This data set proposes a practical approach for handling and reporting breast resection specimens following neoadjuvant therapy. The comments for each data element clarify terminology, discuss available evidence and highlight areas with limited evidence that need further study. This data set overlaps with, and should be used in conjunction with, the data sets for the reporting of invasive breast carcinoma and surgically removed lymph nodes from patients with breast tumours, as appropriate. Key issues specific to the neoadjuvant setting are included in this paper. The entire data set is freely available on the ICCR website. CONCLUSIONS High-quality, standardised reporting of tumour response and residual disease after neoadjuvant treatment are critical for subsequent management decisions for each patient.
Collapse
Affiliation(s)
- Veerle Bossuyt
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Provenzano
- Department of Histopathology, Addenbrookes Hospital, Cambridge, UK
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fleur Webster
- International Collaboration on Cancer Reporting, Surry Hills, NSW, Australia
| | - Kimberly H Allison
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chau Dang
- Memorial Sloan Kettering Cancer Center, West Harrison, NY, USA
| | - Helenice Gobbi
- Department of Surgical Clinic, Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Sunil R Lakhani
- Centre for Clinical Research, and Pathology Queensland, University of Queensland, Brisbane, Qld, Australia
| | - Takuya Moriya
- Department of Pathology, Kawasaki Medical School, Okayama, Japan
| | - Cecily M Quinn
- Department of Histopathology, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College, Dublin, Ireland
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stuart Schnitt
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - D Mark Sibbering
- University Hospitals of Derby and Burton NHS Trust, Royal Derby Hospital, Derby, UK
| | - Elzbieta Slodkowska
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | | - Ian Ellis
- Department of Histopathology, Nottingham City Hospital, London, UK
| |
Collapse
|
14
|
Li ZY, Wu SN, Lin ZH, Jiang MC, Chen C, Liang RX, Lin WJ, Xue ES. Ultrasound-based radiomics-clinical nomogram for noninvasive prediction of residual cancer burden grading in breast cancer. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:566-574. [PMID: 38538081 DOI: 10.1002/jcu.23666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/12/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE To assess the predictive value of an ultrasound-based radiomics-clinical nomogram for grading residual cancer burden (RCB) in breast cancer patients. METHODS This retrospective study of breast cancer patients who underwent neoadjuvant therapy (NAC) and ultrasound scanning between November 2020 and July 2023. First, a radiomics model was established based on ultrasound images. Subsequently, multivariate LR (logistic regression) analysis incorporating both radiomic scores and clinical factors was performed to construct a nomogram. Finally, Receiver operating characteristics (ROC) curve analysis and decision curve analysis (DCA) were employed to evaluate and validate the diagnostic accuracy and effectiveness of the nomogram. RESULTS A total of 1122 patients were included in this study. Among them, 427 patients exhibited a favorable response to NAC chemotherapy, while 695 patients demonstrated a poor response to NAC therapy. The radiomics model achieved an AUC value of 0.84 in the training cohort and 0.83 in the validation cohort. The ultrasound-based radiomics-clinical nomogram achieved an AUC value of 0.90 in the training cohort and 0.91 in the validation cohort. CONCLUSIONS Ultrasound-based radiomics-clinical nomogram can accurately predict the effectiveness of NAC therapy by predicting RCB grading in breast cancer patients.
Collapse
Affiliation(s)
- Zhi-Yong Li
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, China
| | - Sheng-Nan Wu
- Department of Ultrasound, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Ultrasound, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhen-Hu Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mei-Chen Jiang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Cong Chen
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rong-Xi Liang
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wen-Jin Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, China
| | - En-Sheng Xue
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
15
|
Pusztai L, Denkert C, O'Shaughnessy J, Cortes J, Dent R, McArthur H, Kümmel S, Bergh J, Park YH, Hui R, Harbeck N, Takahashi M, Untch M, Fasching PA, Cardoso F, Zhu Y, Pan W, Tryfonidis K, Schmid P. Event-free survival by residual cancer burden with pembrolizumab in early-stage TNBC: exploratory analysis from KEYNOTE-522. Ann Oncol 2024; 35:429-436. [PMID: 38369015 DOI: 10.1016/j.annonc.2024.02.002] [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: 11/30/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND KEYNOTE-522 demonstrated statistically significant improvements in pathological complete response (pCR) with neoadjuvant pembrolizumab plus chemotherapy and event-free survival (EFS) with neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab in patients with high-risk, early-stage triple-negative breast cancer (TNBC). Prior studies have shown the prognostic value of the residual cancer burden (RCB) index to quantify the extent of residual disease after neoadjuvant chemotherapy. In this preplanned exploratory analysis, we assessed RCB distribution and EFS within RCB categories by treatment group. PATIENTS AND METHODS A total of 1174 patients with stage T1c/N1-2 or T2-4/N0-2 TNBC were randomized 2 : 1 to pembrolizumab 200 mg or placebo every 3 weeks given with four cycles of paclitaxel + carboplatin, followed by four cycles of doxorubicin or epirubicin + cyclophosphamide. After surgery, patients received pembrolizumab or placebo for nine cycles or until recurrence or unacceptable toxicity. Primary endpoints are pCR and EFS. RCB is a prespecified exploratory endpoint. The association between EFS and RCB was assessed using a Cox regression model. RESULTS Pembrolizumab shifted patients into lower RCB categories across the entire spectrum compared with placebo. There were more patients in the pembrolizumab group with RCB-0 (pCR), and fewer patients in the pembrolizumab group with RCB-1, RCB-2, and RCB-3. The corresponding hazard ratios (95% confidence intervals) for EFS were 0.70 (0.38-1.31), 0.92 (0.39-2.20), 0.52 (0.32-0.82), and 1.24 (0.69-2.23). The most common first EFS events were distant recurrences, with fewer in the pembrolizumab group across all RCB categories. Among patients with RCB-0/1, more than half [21/38 (55.3%)] of all events were central nervous system recurrences, with 13/22 (59.1%) in the pembrolizumab group and 8/16 (50.0%) in the placebo group. CONCLUSIONS Addition of pembrolizumab to chemotherapy resulted in fewer EFS events in the RCB-0, RCB-1, and RCB-2 categories, with the greatest benefit in RCB-2. These findings demonstrate that pembrolizumab not only increased pCR rates, but also improved EFS among most patients who do not have a pCR.
Collapse
MESH Headings
- Humans
- Female
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/mortality
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Neoplasm, Residual/pathology
- Middle Aged
- Paclitaxel/administration & dosage
- Paclitaxel/therapeutic use
- Paclitaxel/adverse effects
- Carboplatin/administration & dosage
- Neoadjuvant Therapy/methods
- Neoplasm Staging
- Cyclophosphamide/administration & dosage
- Cyclophosphamide/therapeutic use
- Cyclophosphamide/adverse effects
- Aged
- Adult
- Doxorubicin/therapeutic use
- Doxorubicin/administration & dosage
- Epirubicin/administration & dosage
- Epirubicin/therapeutic use
- Progression-Free Survival
- Chemotherapy, Adjuvant/methods
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Double-Blind Method
Collapse
Affiliation(s)
- L Pusztai
- Yale School of Medicine, Yale Cancer Center, New Haven, USA.
| | - C Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg, Marburg, Germany
| | - J O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, US Oncology Network, Dallas, USA
| | - J Cortes
- International Breast Cancer Center, Quironsalud Group, Barcelona; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - R Dent
- National Cancer Center Singapore, Duke - National University of Singapore Medical School, Singapore, Singapore
| | - H McArthur
- University of Texas Southwestern Medical Center, Dallas, USA
| | - S Kümmel
- Breast Unit, Kliniken Essen-Mitte, Essen; Charité - Universitätsmedizin Berlin, Department of Gynecology with Breast Center, Berlin, Germany
| | - J Bergh
- Department of Oncology-Pathology, Karolinska Institutet and Breast Cancer Centre, Cancer Theme, Karolinska University Hospital, Karolinska Comprehensive Cancer Center, Solna, Sweden
| | - Y H Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - R Hui
- Westmead Breast Cancer Institute, Westmead Hospital and the University of Sydney, Sydney, Australia
| | - N Harbeck
- Breast Center, Department of Obstetrics and Gynaecology, LMU University Hospital, Munich, Germany
| | - M Takahashi
- Hokkaido University Hospital, Sapporo, Japan
| | - M Untch
- Breast Cancer Center, Helios Klinikum Berlin-Buch, Berlin
| | - P A Fasching
- University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - F Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal
| | - Y Zhu
- Oncology, Merck & Co., Inc., Rahway, USA
| | - W Pan
- Oncology, Merck & Co., Inc., Rahway, USA
| | | | - P Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
16
|
Tinterri C, Fernandes B, Zambelli A, Sagona A, Barbieri E, Di Maria Grimaldi S, Darwish SS, Jacobs F, De Carlo C, Iuzzolino M, Gentile D. The Impact of Different Patterns of Residual Disease on Long-Term Oncological Outcomes in Breast Cancer Patients Treated with Neo-Adjuvant Chemotherapy. Cancers (Basel) 2024; 16:376. [PMID: 38254865 PMCID: PMC10814808 DOI: 10.3390/cancers16020376] [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: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUNDS The majority of breast cancer (BC) patients treated with neo-adjuvant chemotherapy (NAC) achieves a pathologic partial response with different patterns of residual disease. No clear correlation between these patterns and oncological results was described. Our aims were to define the predictive factors for different patterns of residual disease and compare the outcomes between the scattered versus the circumscribed pattern. METHODS We reviewed 219 postoperative surgical specimens. Patients were divided into two groups: scattered versus circumscribed. Disease-free survival (DFS), distant DFS (DDFS), and overall survival (OS) were analyzed. RESULTS The scattered and circumscribed patterns were assessed in 111 (50.7%) and 108 (49.3%) patients. Two independent predictive factors for the circumscribed pattern were identified: discontinuation of NAC cycles (p = 0.011), and tumor size post-NAC >18 mm (p = 0.022). No difference was observed in terms of DFS and DDFS. Patients with the scattered pattern exhibited a statistically significant better OS. Discontinuation of NAC cycles, tumor size >18 mm, triple-negative BC, and ypN+ were associated with increased recurrence and poorer survival. CONCLUSIONS Discontinuation of NAC cycles and tumor size are independent factors associated with patterns of residual disease. The scattered pattern presents better survival. Understanding the relationship between NAC, the residual pattern, and differences in survival outcomes offers the potential to optimize the therapeutic approaches.
Collapse
Affiliation(s)
- Corrado Tinterri
- Breast Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Bethania Fernandes
- Department of Pathology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Andrea Sagona
- Breast Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Erika Barbieri
- Breast Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | | | - Shadya Sara Darwish
- Breast Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Flavia Jacobs
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Camilla De Carlo
- Department of Pathology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Martina Iuzzolino
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
- Department of Pathology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Damiano Gentile
- Breast Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| |
Collapse
|
17
|
Miglietta F, Ragazzi M, Fernandes B, Griguolo G, Massa D, Girardi F, Bottosso M, Bisagni A, Zarrilli G, Porra F, Iannaccone D, Dore L, Gaudio M, Santandrea G, Fassan M, Lo Mele M, De Sanctis R, Zambelli A, Bisagni G, Guarneri V, Dieci MV. A Prognostic Model Based on Residual Cancer Burden and Tumor-Infiltrating Lymphocytes on Residual Disease after Neoadjuvant Therapy in HER2+ Breast Cancer. Clin Cancer Res 2023; 29:3429-3437. [PMID: 37417941 PMCID: PMC10472099 DOI: 10.1158/1078-0432.ccr-23-0480] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/26/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
PURPOSE We aim to evaluate the prognostic significance of tumor-infiltrating lymphocyte on residual disease (RD-TIL) in HER2+ patients with breast cancer who failed to achieve pathologic complete response (pCR) after anti-HER2+ chemotherapy (CT)-based neoadjuvant treatment (NAT). We assessed the feasibility of combining the prognostic information provided by residual cancer burden (RCB) and RD-TILs into a composite score (RCB+TIL). EXPERIMENTAL DESIGN HER2+ patients with breast cancer treated with CT+anti-HER2-based NAT at three institutions were retrospectively included. RCB and TIL levels were evaluated on hematoxylin and eosin-stained slides from surgical samples according to available recommendations. Overall survival (OS) was used as an outcome measure. RESULTS A total of 295 patients were included, of whom 195 had RD. RCB was significantly associated with OS. Higher RD-TILs were significantly associated with poorer OS as compared with lower RD-TILs (15% cutoff). In multivariate analysis, both RCB and RD-TIL maintained their independent prognostic value. A combined score, RCB+TIL, was calculated from the estimated coefficient of RD-TILs and the RCB index in a bivariate logistic model for OS. The RCB+TIL score was significantly associated with OS. The C-index for OS of the RCB+TIL score was numerically higher than that of RCB and significantly higher than that of RD-TILs. CONCLUSIONS We have reported an independent prognostic impact of RD-TILs after anti-HER2+CT NAT, which might underlie an imbalance of the RD microenvironment towards immunosuppressive features. We provided a new composite prognostic score based on RCB+TIL, which was significantly associated with OS and proved to be more informative than the isolated evaluation of RCB and RD-TILs.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Moira Ragazzi
- Pathology Unit, Arcispedale Santa Maria Nuova, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Davide Massa
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Fabio Girardi
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Michele Bottosso
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Alessandra Bisagni
- Pathology Unit, Arcispedale Santa Maria Nuova, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giovanni Zarrilli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy
| | - Francesca Porra
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Daniela Iannaccone
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Leocadia Dore
- Humanitas Clinical and Research Center – IRCCS, Rozzano (MI), Italy
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Mariangela Gaudio
- Humanitas Clinical and Research Center – IRCCS, Rozzano (MI), Italy
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Giacomo Santandrea
- Pathology Unit, Arcispedale Santa Maria Nuova, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Fassan
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy
| | - Marcello Lo Mele
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy
| | - Rita De Sanctis
- Humanitas Clinical and Research Center – IRCCS, Rozzano (MI), Italy
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Alberto Zambelli
- Humanitas Clinical and Research Center – IRCCS, Rozzano (MI), Italy
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Giancarlo Bisagni
- Oncology Unit, Arcispedale Santa Maria Nuova, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto – IOV IRCCS, Padova, Italy
| |
Collapse
|
18
|
Oner G, Broeckx G, Van Berckelaer C, Zwaenepoel K, Altintas S, Canturk Z, Tjalma W, Berneman Z, Peeters M, Pauwels P, van Dam PA. The immune microenvironment characterisation and dynamics in hormone receptor-positive breast cancer before and after neoadjuvant endocrine therapy. Cancer Med 2023; 12:17901-17913. [PMID: 37553911 PMCID: PMC10524081 DOI: 10.1002/cam4.6425] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 07/05/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Oestrogen receptor positive (ER+)/HER-2 negative breast cancer (BC) is considered to be an immunologically cold tumour compared to triple negative breast cancer. Therefore, the tumour microenvironment (TME) of ER+/HER-2 negative BC is understudied. The aim of this project is to investigate the TME and the immune response during neoadjuvant endocrine therapy (NET) and to correlate this with the treatment response in a real life setting. METHODS Expression of immune checkpoint receptors and immune cells was examined immunohistochemically, pre- and post-NET in a cohort of 56 ER+/HER-2 negative BC patients. They were treated with tamoxifen (n = 16), an aromatase inhibitor (n = 40) or a combination of an aromatase inhibitor with a PI3K inhibitor (n = 11) for a median duration of 6 months (range 1-32 months). Immunohistochemical staining with monoclonal antibodies for PDL-1, PD-1, TIM-3, LAG-3, CTLA-4, CD4, CD68 and FOXP3 were performed. All staining procedures were done according to validated protocols, and scoring was done by a pathologist specialized in breast cancer. Positivity was defined as staining >1% on TILs. Response to NET was evaluated according to tumour size change on imaging and Ki-67 change. RESULTS The median age was 61.02 (37-90) years. Diameter of tumour size decreased with a mean of 8.1 mm (-16 mm to 45 mm) (p < 0.001) during NET and the value of Ki-67 value decreased with a median of 9 after NET (p < 0.001). An increase in PD-L1 expression after NET showed a trend towards significant (p = 0.088) and CD-4+ T cells significantly increased after NET (p = 0.03). A good response to NET defined as a decrease in tumour size and/or decrease of Ki-67 was found to be associated with a longer duration of NET, a change of CD4+ T-cells and a higher number of CD68+ tumour-associated macrophages before the start of NET. CONCLUSION The immune microenvironment plays an important role in ER+/HER-2 negative BC. NET influences the composition and functional state of the infiltrating immune cells. Furthermore, changes in the immune microenvironment are also associated with treatment response.
Collapse
Affiliation(s)
- Gizem Oner
- Multidisciplinary Oncologic Centre Antwerp (MOCA)Antwerp University HospitalEdegemBelgium
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of General SurgeryKocaeli UniversityKocaeliTurkey
| | - Glenn Broeckx
- Department of HistopathologyAntwerp University HospitalEdegemBelgium
| | | | - Karen Zwaenepoel
- Department of HistopathologyAntwerp University HospitalEdegemBelgium
| | - Sevilay Altintas
- Multidisciplinary Oncologic Centre Antwerp (MOCA)Antwerp University HospitalEdegemBelgium
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| | - Zafer Canturk
- Department of General SurgeryKocaeli UniversityKocaeliTurkey
| | - Wiebren Tjalma
- Multidisciplinary Oncologic Centre Antwerp (MOCA)Antwerp University HospitalEdegemBelgium
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| | - Zwi Berneman
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of HematologyAntwerp University HospitalEdegemBelgium
| | - Marc Peeters
- Multidisciplinary Oncologic Centre Antwerp (MOCA)Antwerp University HospitalEdegemBelgium
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
- Department of HistopathologyAntwerp University HospitalEdegemBelgium
| | - Peter A. van Dam
- Multidisciplinary Oncologic Centre Antwerp (MOCA)Antwerp University HospitalEdegemBelgium
- Center for Oncological Research (CORE)University of AntwerpWilrijkBelgium
| |
Collapse
|
19
|
Lejeune M, Reverté L, Gallardo N, Sauras E, Bosch R, Mata D, Roso A, Petit A, Peg V, Riu F, García-Fontgivell J, Relea F, Vieites B, de la Cruz-Merino L, Arenas M, Rodriguez V, Galera J, Korzynska A, Plancoulaine B, Álvaro T, López C. Matrix Metalloproteinase-9 Expression Is Associated with the Absence of Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer Patients. Int J Mol Sci 2023; 24:11297. [PMID: 37511057 PMCID: PMC10378773 DOI: 10.3390/ijms241411297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is particularly challenging due to the weak or absent response to therapeutics and its poor prognosis. The effectiveness of neoadjuvant chemotherapy (NAC) response is strongly influenced by changes in elements of the tumor microenvironment (TME). This work aimed to characterize the residual TME composition in 96 TNBC patients using immunohistochemistry and in situ hybridization techniques and evaluate its prognostic implications for partial responders vs. non-responders. Compared with non-responders, partial responders containing higher levels of CD83+ mature dendritic cells, FOXP3+ regulatory T cells, and IL-15 expression but lower CD138+ cell concentration exhibited better OS and RFS. However, along with tumor diameter and positive nodal status at diagnosis, matrix metalloproteinase-9 (MMP-9) expression in the residual TME was identified as an independent factor associated with the impaired response to NAC. This study yields new insights into the key components of the residual tumor bed, such as MMP-9, which is strictly associated with the lack of a pathological response to NAC. This knowledge might help early identification of TNBC patients less likely to respond to NAC and allow the establishment of new therapeutic targets.
Collapse
Affiliation(s)
- Marylène Lejeune
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Laia Reverté
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Noèlia Gallardo
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Esther Sauras
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
- Clinical Studies Unit, Hospital de Tortosa Verge de la Cinta, Carretera Esplanetes, 14, 43500 Tortosa, Spain
| | - Ramon Bosch
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Daniel Mata
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Albert Roso
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Gran Via Corts Catalanes, 587, 08007 Barcelona, Spain
| | - Anna Petit
- Pathology Department, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Vicente Peg
- Pathology Department, Hospital Universitari de Vall Hebron, 08035 Barcelona, Spain
| | - Francisco Riu
- Pathology Department, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Joan García-Fontgivell
- Pathology Department, Hospital Universitari Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Fernanda Relea
- Pathology Department, Hospital General de Ciudad Real, 13005 Ciudad Real, Spain
| | - Begoña Vieites
- Pathology Department, Hospital Universitario Virgen del Rocío, 41013 Seville, Spain
| | | | - Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Tarragona, Spain
| | - Valeri Rodriguez
- Oncology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), 43500 Tortosa, Spain
| | - Juana Galera
- Gynaecology Department, Hospital Universitari Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Anna Korzynska
- Laboratory of Processing and Analysis of Microscopic Images, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Benoît Plancoulaine
- ANTICIPE, INSERM, François Baclesse Comprehensive Cancer Center, University Caen Normandy, 14000 Caen, France
| | - Tomás Álvaro
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Carlos López
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| |
Collapse
|
20
|
Sejben A, Hegedűs F, Almási S, Berta M, Oláh-Németh O, Zombori T. Good practice: The experiences with the utilization of residual cancer burden-A single institution study. Thorac Cancer 2023; 14:963-968. [PMID: 36866777 PMCID: PMC10101829 DOI: 10.1111/1759-7714.14826] [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: 12/13/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
INTRODUCTION The use of neoadjuvant therapy (NAT) has been showing an incraesing tendency in the treatment of locally advanced breast cancer. The evaluation of residual cancer could be performed by Residual Cancer Burden (RCB) calculator. The prognostic system takes the two largest diameters of the tumor, the cellularity, the amount of in situ carcinoma, the number of metastatic lymph nodes, and the size of the largest metastatic deposit into account. The aim of our study was to examine the reproducibility of RCB in NAT treated patients. METHODS Patients who were treated with NAT and had resection specimens between 2018 and 2021 were selected. Histological examination was performed by five pathologists. After assessment of the examined variables, RCB points and RCB classes were defined. For statistical analysis, interclass correlation was used (SPSS Statistics V.22.0 software). RESULTS Altogether 100 patients were included in our retrospective, cohort study (average age: 57 years). In two-thirds of the cases, third generation chemotherapy was used, and mastectomy was performed. Significant concordance was found in the two largest diameters of the tumor (coefficients, 0.984 and 0.973), the cellularity (coefficient, 0.970), and the largest metastatic deposit (coefficient, 0.998). Although the amount of in situ carcinoma proved to be the least reproducible factor, it resulted in almost 90% of agreement (coefficient, 0.873). Regarding RCB points and classes, similar results were observed (coefficients, 0.989 and 0.960). CONCLUSIONS Significant agreement was observed between examiners based on almost all RCB parameters, points, and classes, reflecting the optimal reproducibility of RCB. Therefore, we recommend the use of the calculator in routine histopathological reports in NAT cases.
Collapse
Affiliation(s)
- Anita Sejben
- Department of Pathology, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Fanni Hegedűs
- Department of Pathology, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Szintia Almási
- Department of Pathology, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Márton Berta
- Department of Pathology, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Orsolya Oláh-Németh
- Department of Pathology, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Tamás Zombori
- Department of Pathology, University of Szeged, Faculty of Medicine, Szeged, Hungary
| |
Collapse
|
21
|
Wang M, Ding Q, Gu J, Sfamenos SM, Huo L, Tang Z, Sun H, Robinson M, Tang G, Lim B, Wu Y, Albarracin CT, Sahin AA, Chen H. Breast Cancer With a HER2 FISH Group 2 Result: Should HER2 Tests be Repeated? Clin Breast Cancer 2023; 23:415-422. [PMID: 36878823 DOI: 10.1016/j.clbc.2023.02.006] [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: 12/21/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Breast cancer with fluorescence in situ hybridization (FISH) group 2 pattern (HER2 <4 and HER2/CEP17 ratio ≥2, a subset of monosomy CEP17) was historically considered HER2-positive, but mostly HER2-negative according to updated 2018 American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guidelines unless 3+ by immunohistochemistry (IHC). Therapeutic relevance of this group remained elusive, therefore we assessed if repeat IHC and FISH can assist final HER2 classification. PATIENT AND METHODS We retrospectively reviewed HER2 FISH performed at our institution from 2014 to 2018 and identified 23 of 3554 (0.6%) breast cancer cases with at least one-time measurement of HER2 FISH categorized as group 2. Repeat HER2 tests were performed for cases with available alternative tumor samples and compared with initial testing following 2018 ASCO/CAP guidelines. RESULTS Only 1 of 23 group 2 cases was HER2-positive, 0/18 in primary and 1/5 in metastatic/recurrent tumors. Of 13 primary tumors with repeat HER2 results; 10 (77%) remained HER2-negative; 3 (23%) changed from HER2-negative (group 2 and IHC 2+) to HER2-positive (group 1 and IHC 2+). Among 8 of these 13 patients receiving neoadjuvant systemic therapy containing anti-HER2 agent, 3 (38%) achieved pathologic complete response (pCR). Two of 3 pCR cases were HER2-positive converters on repeat testing. Three pCR cases were ER-negative or -low positive and Ki67 ≥40%, while 5 partial responders were ER-positive and Ki67 <40% (P < .05). CONCLUSION Breast cancer with HER2 FISH group 2 result may represent heterogeneous populations of tumor cells being originated de novo or preferentially selected secondary to therapy. Repeat HER2 tests on alternative samples may be considered to guide anti-HER2 therapy.
Collapse
Affiliation(s)
- Minhua Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qingqing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jun Gu
- The University of Texas MD Anderson Cancer Center, School of Health Professions, Houston, TX
| | - Steven M Sfamenos
- The University of Texas MD Anderson Cancer Center, School of Health Professions, Houston, TX
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zhenya Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hongxia Sun
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Melissa Robinson
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yun Wu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Aysegul A Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hui Chen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
22
|
da Silva JL, Carvalho GDS, Zanetti de Albuquerque L, Rodrigues FR, Fernandes PV, Kischinhevsky D, de Melo AC. Exploring Real-World HER2-Low Data in Early-Stage Triple-Negative Breast Cancer: Insights and Implications. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:337-347. [PMID: 37188066 PMCID: PMC10178312 DOI: 10.2147/bctt.s408743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/30/2023] [Indexed: 05/17/2023]
Abstract
Purpose This study aimed to compare the clinical behavior, clinicopathological and sociodemographic characteristics of patients with early-stage triple-negative breast cancer (TNBC) who belong to the HER2-low and HER2-zero subgroups. Patients and Methods This study involved a thorough search in the internal database of a single Brazilian institution to identify women with TNBC who underwent neoadjuvant chemotherapy (NACT) followed by curative surgery within the period from January 2010 to December 2014. HER2 analysis through immunohistochemistry (IHC) and, if required, amplification by in situ hybridization, was conducted using core biopsy samples. The study assesses outcomes of residual cancer burden (RCB), event-free survival (EFS), and overall survival (OS). Results A total of 170 cases were analyzed, with a mean age of 51.4 years (standard deviation, SD 11.2). The HER2 status was categorized as IHC 0, 1+, or 2+ in 80 (47.1%), 73 (42.9%), and 17 (10%) patients, respectively. No significant differences were observed in the prevalence of clinical pathological characteristics among the subgroups. The absence of significant results for clinicopathological and demographic features hindered the multivariate analysis of HER2 subgroups. Similarly, no significant differences were found in the RCB, EFS, and OS outcomes between HER2 subgroups. Conclusion The findings of this study suggest that, in early-stage TNBC, the clinical behavior and survival outcomes of the HER2-low subgroup may not differ significantly from those of the HER2-zero subgroup.
Collapse
Affiliation(s)
- Jesse Lopes da Silva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
- Correspondence: Jesse Lopes da Silva, Brazilian National Cancer Institute (INCA), Clinical Research Division, 37 André Cavalcanti Street, 5th Floor, Annex Building, Rio de Janeiro, 20231-050, Brazil, Tel/Fax +55 21 32076585, Email
| | - Giselle de Souza Carvalho
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Lucas Zanetti de Albuquerque
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | | | | | - Daniel Kischinhevsky
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Andreia Cristina de Melo
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Pestana CV, Livasy CA, Donahue EE, Neelands B, Tan AR, Sarantou T, Hadzikadic-Gusic L, White RL. Does Residual Cancer Burden Predict Local Recurrence After Neoadjuvant Chemotherapy? Ann Surg Oncol 2022; 29:7716-7724. [PMID: 35810226 DOI: 10.1245/s10434-022-12038-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/30/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND The extent of residual disease after neoadjuvant chemotherapy (NAC) can be quantified by the Residual Cancer Burden (RCB), a prognostic tool used to estimate survival outcomes in breast cancer. This study investigated the association between RCB and locoregional recurrence (LRR). METHODS The study reviewed 532 women with breast cancer who underwent NAC between 2010 and 2016. Relapse in the ipsilateral breast, skin/subcutis at the surgical site, chest wall, pectoralis, or regional lymph nodes defined an LRR. The LRR cumulative incidence (LRCI) was estimated using the Fine and Gray competing-risks model, with death and distant recurrence defined as competing events. The association of LRCI with prognostic variables was evaluated. RESULTS Overall, 5.5% of the patients experienced an LRR after a median follow-up period of 65 months. The 5-year LRCI rates by RCB were as follows: RCB-0 (0.9%), RCB-1 (3.2%), RCB-2 (6.0%), and RCB-3 (12.9%). In the univariable analysis, LRCI varied significantly by RCB (p = 0.010). The multivariable analysis showed a significant association of LRCI with increasing RCB, and the patients with hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) phenotype were at lower risk for LRR than those with HER2+ and triple-negative cancers (p < 0.032). The patients with RCB-3 were at a higher risk for local relapse than those with RCB-0 (hazard ratio, 13.78; confidence interval, 2.25-84.45; p = 0.04). Type of operation (p = 0.04) and use of adjuvant radiation (p = 0.046) were statistically significant in the multivariable model. CONCLUSIONS The study results demonstrate a significant association between LRCI and increasing RCB, although distant recurrence is a substantial driver of disease outcomes. Future prospective studies should examine the role of RCB in clinical decisions regarding indications for adjuvant therapy.
Collapse
Affiliation(s)
- Christine V Pestana
- Division of Surgical Oncology, Department of Surgery, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Chad A Livasy
- Department of Pathology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Erin E Donahue
- Department of Cancer Biostatistics, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Brittany Neelands
- Division of Surgical Oncology, Department of Surgery, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Antoinette R Tan
- Department of Solid Tumor and Investigational Therapeutics, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Terry Sarantou
- Division of Surgical Oncology, Department of Surgery, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Lejla Hadzikadic-Gusic
- Division of Surgical Oncology, Department of Surgery, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Richard L White
- Division of Surgical Oncology, Department of Surgery, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA.
| |
Collapse
|
24
|
Atzori G, Gipponi M, Cornacchia C, Diaz R, Sparavigna M, Gallo M, Ruelle T, Murelli F, Franchelli S, Depaoli F, Friedman D, Fregatti P. " No Ink on Tumor" in Breast-Conserving Surgery after Neoadjuvant Chemotherapy. J Pers Med 2022; 12:jpm12071031. [PMID: 35887526 PMCID: PMC9320436 DOI: 10.3390/jpm12071031] [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: 05/27/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Background/Aim: Patients with Stage I-II breast cancer undergoing breast-conserving surgery after neoadjuvant chemotherapy (BCS-NAC) were retrospectively assessed in order to evaluate the extent of a safe excision margin. Materials and Methods: Between 2003 and 2020, 151 patients underwent risk-adapted BCS-NAC; margin involvement was always assessed at definitive histology. Patients with complete pathological response (pCR) were classified as the RX group, whereas those with residual disease and negative margins were stratified as R0 < 1 mm (margin < 1 mm) and R0 > 1 mm (margin > 1 mm). Results: Totals of 29 (19.2%), 64 (42.4%), and 58 patients (38.4%) were included in the R0 < 1 mm, R0 > 1 mm, and RX groups, respectively, and 2 patients with margin involvement had a mastectomy. Ten instances of local recurrence (6.6%) occurred, with no statistically significant difference in local recurrence-free survival (LRFS) between the three groups. A statistically significant advantage of disease-free survival (p = 0.002) and overall survival (p = 0.010) was observed in patients with pCR. Conclusions: BCS-NAC was increased, especially in HER-2-positive and triple-negative tumors; risk-adapted BCS should be preferably pursued to highlight the cosmetic benefit of NAC. The similar rate of LRFS in the three groups of patients suggests a shift toward the “no ink on tumor” paradigm for patients undergoing BCS-NAC.
Collapse
Affiliation(s)
- Giulia Atzori
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
| | - Marco Gipponi
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
- Correspondence: ; Tel.: +30-010-5558805
| | - Chiara Cornacchia
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
| | - Raquel Diaz
- Department Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, 16132 Genoa, Italy; (R.D.); (T.R.)
| | - Marco Sparavigna
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
| | - Maurizio Gallo
- Department of Internal Medicine (Di.M.I.), University of Genoa, 16132 Genoa, Italy;
| | - Tommaso Ruelle
- Department Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, 16132 Genoa, Italy; (R.D.); (T.R.)
| | - Federica Murelli
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
- Department Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, 16132 Genoa, Italy; (R.D.); (T.R.)
| | - Simonetta Franchelli
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
| | - Francesca Depaoli
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
| | - Daniele Friedman
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
- Department Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, 16132 Genoa, Italy; (R.D.); (T.R.)
| | - Piero Fregatti
- Breast Surgery Clinic, San Martino Policlinic Hospital, 16132 Genoa, Italy; (G.A.); (C.C.); (M.S.); (F.M.); (S.F.); (F.D.); (D.F.); (P.F.)
- Department Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, 16132 Genoa, Italy; (R.D.); (T.R.)
| |
Collapse
|
25
|
The association between breast density and breast cancer pathological response to neoadjuvant chemotherapy. Breast Cancer Res Treat 2022; 194:385-392. [PMID: 35606616 PMCID: PMC9239960 DOI: 10.1007/s10549-022-06616-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 04/30/2022] [Indexed: 11/21/2022]
Abstract
Purpose Mammographic Density (MD) refers to the amount of fibroglandular breast tissue present in the breast and is an established risk factor for developing breast cancer. The ability to evaluate treatment response dynamically renders neoadjuvant chemotherapy (NACT) the preferred treatment option in many clinical scenarios. Previous studies have suggested that MD can predict patients likely to achieve a pathological complete response (pCR) to NACT. We aimed to determine whether there is a causal relationship between BI-RADS breast composition categories for breast density at diagnosis and the pCR rate and residual cancer burden score (RCB) by performing a retrospective review on consecutive breast cancer patients who received NACT in a tertiary referral centre from 2015 to 2021. Methods The Mann–Whitney U Test was used to test for differences between two independent groups (i.e. those who achieved pCR and those who did not). A binary logistic regression model was used to estimate odds ratios (OR) and corresponding 95% confidence intervals (CI) for an association between the independent variables of molecular subtype, MD, histological grade and FNA positivity and the dependant variable of pCR. Statistical analysis was conducted with SPSS (IBM SPSS for Mac, Version 26.0; IBM Corp). Results 292 patients were included in the current study. There were 124, 155 and 13 patients in the BI-RADS MD category b, c and d, respectively. There were no patients in the BI-RADS MD category a. The patients with less dense breast composition (MD category b) were significantly older than patients with denser breast composition (MD category c, d) (p = 0.001) and patients who had a denser breast composition (MD category d) were more likely to have ER+ tumours. There was no significant difference in PgR status, HER2 status, pathological complete response (pCR), FNA positivity, or RCB class dependent upon the three MD categories. A binary logistic regression revealed that patients with HER2-enriched breast cancer and triple-negative breast cancer are more likely to achieve pCR with an OR of 3.630 (95% CI 1.360–9.691, p = 0.010) and 2.445 (95% CI 1.131–5.288, p = 0.023), respectively. Conclusion Whilst dense MD was associated with ER positivity and these women were less likely to achieve a pCR, MD did not appear to independently predict pCR post-NACT.
Collapse
|
26
|
Han D, Liao J, Zhang M, Qin C, Han M, Wu C, Li J, Yao J, Liu Y. Reconstructing virtual large slides can improve the accuracy and consistency of tumor bed evaluation for breast cancer after neoadjuvant therapy. Diagn Pathol 2022; 17:40. [PMID: 35484579 PMCID: PMC9047297 DOI: 10.1186/s13000-022-01219-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 04/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To explore whether the "WSI Stitcher", a program we developed for reconstructing virtual large slide through whole slide imaging fragments stitching, can improve the efficiency and consistency of pathologists in evaluating the tumor bed after neoadjuvant treatment of breast cancer compared with the conventional methods through stack splicing of physical slides. METHODS This study analyzed the advantages of using software-assisted methods to evaluate the tumor bed after neoadjuvant treatment of breast cancer. This new method is to use "WSI Stitcher" to stitch all the WSI fragments together to reconstruct a virtual large slide and evaluate the tumor bed with the help of the built-in ruler and tumor proportion calculation functions. RESULTS Compared with the conventional method, the evaluation time of the software-assisted method was shortened by 35%(P < 0.001). In the process of tumor bed assessment after neoadjuvant treatment of breast cancer, the software-assisted method has higher intraclass correlation coefficient when measuring the length (0.994 versus 0.934), width (0.992 versus 0.927), percentage of residual tumor cells (0.947 versus 0.878), percentage of carcinoma in situ (0.983 versus 0.881) and RCB index(0.997 versus 0.772). The software-assisted method has higher kappa values when evaluating tumor staging(0.901 versus 0.687) and RCB grading (0.963 versus 0.857). CONCLUSION The "WSI Stitcher" is an effective tool to help pathologists with the assessment of breast cancer after neoadjuvant treatment.
Collapse
Affiliation(s)
- Dandan Han
- Department of Pathology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jun Liao
- AI Lab, Tencent, Tencent Binhai Building, No. 33, Haitian Second Road, Nanshan District, Shenzhen, 518054, Guangdong, China
| | - Meng Zhang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Chenchen Qin
- AI Lab, Tencent, Tencent Binhai Building, No. 33, Haitian Second Road, Nanshan District, Shenzhen, 518054, Guangdong, China
| | - Mengxue Han
- Department of Pathology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Chun Wu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jinze Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jianhua Yao
- AI Lab, Tencent, Tencent Binhai Building, No. 33, Haitian Second Road, Nanshan District, Shenzhen, 518054, Guangdong, China.
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
| |
Collapse
|
27
|
Troxell ML, Gupta T. Neoadjuvant Therapy in Breast Cancer: Histologic Changes and Clinical Implications. Surg Pathol Clin 2022; 15:57-75. [PMID: 35236634 DOI: 10.1016/j.path.2021.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cytotoxic or endocrine therapy before surgery (neoadjuvant) for breast cancer has become standard of care, affording the opportunity to assess and quantify response in the subsequent resection specimen. Correlation with radiology, cassette mapping, and histologic review with a semi-quantitative reporting system such as residual cancer burden (RCB) provides important prognostic data that may guide further therapy. The tumor bed should be identified histologically, often as a collagenized zone devoid of normal breast epithelium, with increased vasculature. Identification of residual treated carcinoma may require careful high power examination, as residual tumor cells may be small and dyscohesive; features are widely variable and include hyperchromatic small, large, or multiple nuclei with clear, foamy, or eosinophilic cytoplasm. Calculation of RCB requires residual carcinoma span in 2 dimensions, estimated carcinoma cellularity (% area), number of involved lymph nodes, and span of largest nodal carcinoma. These RCB parameters may differ from AJCC staging measurements, which depend on only contiguous carcinoma in breast and lymph nodes.
Collapse
Affiliation(s)
- Megan L Troxell
- Department of Pathology, Stanford University School of Medicine, Stanford Pathology, 300 Pasteur Drive, H2110, Stanford, CA 94305, USA.
| | - Tanya Gupta
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, 900 Blake Wilbur Drive, Palo Alto, CA 94304 USA
| |
Collapse
|
28
|
Xiang Y, Chen L, Liu C, Yi X, Li L, Huang Y. Redirecting Chemotherapeutics to the Endoplasmic Reticulum Increases Tumor Immunogenicity and Potentiates Anti-PD-L1 Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104591. [PMID: 34859582 DOI: 10.1002/smll.202104591] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/28/2021] [Indexed: 05/21/2023]
Abstract
The endoplasmic reticulum (ER) in cancer cells has been considered as a pharmacological target. Still, the effects of a ER-targeted system remain less investigated, due to the fact that most chemo-drugs take actions in the nucleus. Here, it is demonstrated that ER-targeted delivery of doxorubicin (DOX), a typically nucleus-tropic-and-acting agent, attenuates its original effect on cytotoxicity while generating new functions favorable for immune activation. First, a library of DOX derivatives with variable ER-targeting abilities is synthesized. The results reveal that higher ER-targeting efficiency correlates with greater ER stress. As compared with naïve drug, ER-targeted DOX considerably alters the mode of action from nuclear DNA damage-associated cytotoxicity to ER stress-mediated calreticulin exposure. Consequently, ER-targeted DOX decreases cytotoxicity but increases the capability to induce immunogenic cell death (ICD). Therefore, a platform combining naïve and ER-targeted DOX is constructed for in vivo application. Conventional polymer-DOX conjugate inhibits tumor growth by exerting a direct killing effect, and ER-targeted polymer-DOX conjugate suppresses residual tumors by eliciting ICD-associated immunity, together resulting in considerable tumor regression. In addition, simultaneous inhibition of adaptive PD-L1 enrichment (due to negative-feedback to ICD induction) further leads to greater therapeutic outcome. Collectively, ER-targeted therapy can enhance anticancer efficacy by promoting ICD-associated immunotherapy, and potentiating chemotherapy and checkpoint blockade therapy.
Collapse
Affiliation(s)
- Yucheng Xiang
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Liqiang Chen
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Chendong Liu
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Xiaoli Yi
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Lian Li
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Yuan Huang
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| |
Collapse
|
29
|
Yau C, Osdoit M, van der Noordaa M, Shad S, Wei J, de Croze D, Hamy AS, Laé M, Reyal F, Sonke GS, Steenbruggen TG, van Seijen M, Wesseling J, Martín M, Del Monte-Millán M, López-Tarruella S, Boughey JC, Goetz MP, Hoskin T, Gould R, Valero V, Edge SB, Abraham JE, Bartlett JMS, Caldas C, Dunn J, Earl H, Hayward L, Hiller L, Provenzano E, Sammut SJ, Thomas JS, Cameron D, Graham A, Hall P, Mackintosh L, Fan F, Godwin AK, Schwensen K, Sharma P, DeMichele AM, Cole K, Pusztai L, Kim MO, van 't Veer LJ, Esserman LJ, Symmans WF. Residual cancer burden after neoadjuvant chemotherapy and long-term survival outcomes in breast cancer: a multicentre pooled analysis of 5161 patients. Lancet Oncol 2022; 23:149-160. [PMID: 34902335 PMCID: PMC9455620 DOI: 10.1016/s1470-2045(21)00589-1] [Citation(s) in RCA: 260] [Impact Index Per Article: 86.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous studies have independently validated the prognostic relevance of residual cancer burden (RCB) after neoadjuvant chemotherapy. We used results from several independent cohorts in a pooled patient-level analysis to evaluate the relationship of RCB with long-term prognosis across different phenotypic subtypes of breast cancer, to assess generalisability in a broad range of practice settings. METHODS In this pooled analysis, 12 institutes and trials in Europe and the USA were identified by personal communications with site investigators. We obtained participant-level RCB results, and data on clinical and pathological stage, tumour subtype and grade, and treatment and follow-up in November, 2019, from patients (aged ≥18 years) with primary stage I-III breast cancer treated with neoadjuvant chemotherapy followed by surgery. We assessed the association between the continuous RCB score and the primary study outcome, event-free survival, using mixed-effects Cox models with the incorporation of random RCB and cohort effects to account for between-study heterogeneity, and stratification to account for differences in baseline hazard across cancer subtypes defined by hormone receptor status and HER2 status. The association was further evaluated within each breast cancer subtype in multivariable analyses incorporating random RCB and cohort effects and adjustments for age and pretreatment clinical T category, nodal status, and tumour grade. Kaplan-Meier estimates of event-free survival at 3, 5, and 10 years were computed for each RCB class within each subtype. FINDINGS We analysed participant-level data from 5161 patients treated with neoadjuvant chemotherapy between Sept 12, 1994, and Feb 11, 2019. Median age was 49 years (IQR 20-80). 1164 event-free survival events occurred during follow-up (median follow-up 56 months [IQR 0-186]). RCB score was prognostic within each breast cancer subtype, with higher RCB score significantly associated with worse event-free survival. The univariable hazard ratio (HR) associated with one unit increase in RCB ranged from 1·55 (95% CI 1·41-1·71) for hormone receptor-positive, HER2-negative patients to 2·16 (1·79-2·61) for the hormone receptor-negative, HER2-positive group (with or without HER2-targeted therapy; p<0·0001 for all subtypes). RCB score remained prognostic for event-free survival in multivariable models adjusted for age, grade, T category, and nodal status at baseline: the adjusted HR ranged from 1·52 (1·36-1·69) in the hormone receptor-positive, HER2-negative group to 2·09 (1·73-2·53) in the hormone receptor-negative, HER2-positive group (p<0·0001 for all subtypes). INTERPRETATION RCB score and class were independently prognostic in all subtypes of breast cancer, and generalisable to multiple practice settings. Although variability in hormone receptor subtype definitions and treatment across patients are likely to affect prognostic performance, the association we observed between RCB and a patient's residual risk suggests that prospective evaluation of RCB could be considered to become part of standard pathology reporting after neoadjuvant therapy. FUNDING National Cancer Institute at the US National Institutes of Health.
Collapse
Affiliation(s)
- Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
| | - Marie Osdoit
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Surgery, Institut Curie, Paris, France
| | | | - Sonal Shad
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jane Wei
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Diane de Croze
- Department of Tumor Biology, Institut Curie, Paris, France
| | | | - Marick Laé
- Department of Tumor Biology, Institut Curie, Paris, France; Department of Pathology, Université de Rouen Normandie, Rouen, France
| | - Fabien Reyal
- Department of Surgery, Institut Curie, Paris, France
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Tessa G Steenbruggen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maartje van Seijen
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jelle Wesseling
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Miguel Martín
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Maria Del Monte-Millán
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Sara López-Tarruella
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | | | - Tanya Hoskin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Rebekah Gould
- Department of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen B Edge
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jean E Abraham
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - John M S Bartlett
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, Canada; Deanery of Molecular, Genetic and Population Health Sciences, Edinburgh Cancer Research Centre, Edinburgh, UK; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Carlos Caldas
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Janet Dunn
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Helena Earl
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Larry Hayward
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Louise Hiller
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Elena Provenzano
- Department of Histopathology, University of Cambridge, Cambridge, UK
| | | | - Jeremy S Thomas
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - David Cameron
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Ashley Graham
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Peter Hall
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Lorna Mackintosh
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Fang Fan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kelsey Schwensen
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Priyanka Sharma
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Angela M DeMichele
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly Cole
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Lajos Pusztai
- Department of Medical Oncology, Yale University, New Haven, CT, USA
| | - Mi-Ok Kim
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - W Fraser Symmans
- Department of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
30
|
Wang W, Liu Y, Zhang H, Zhang S, Duan X, Ye J, Xu L, Zhao J, Cheng Y, Liu Q. Prognostic value of residual cancer burden and Miller-Payne system after neoadjuvant chemotherapy for breast cancer. Gland Surg 2021; 10:3211-3221. [PMID: 35070881 PMCID: PMC8749085 DOI: 10.21037/gs-21-608] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/05/2021] [Indexed: 03/12/2024]
Abstract
BACKGROUND To verify the feasibility of using the residual cancer burden (RCB) index to stratify prognosis of patients after neoadjuvant chemotherapy (NAC) and to compare RCB with the Miller-Payne system. METHODS We retrospectively analyzed clinicopathological data of patients receiving treatment between January 1, 2010 and December 31, 2018. Kaplan-Meier curves were used to compare the survival outcomes and estimate disease-free survival (DFS) and disease-specific survival (DSS). Harrell's concordance index (C-index) was used to evaluate the predictive accuracy of RCB and Miller-Payne system. RESULTS A total of 423 female patients with complete data were included in the analysis, with a median follow-up time of 58.5 months (range, 7-126 months); 84 patients experienced recurrence, and 48 experienced breast cancer related death. RCB index and the Miller-Payne system were associated with prognosis in the whole cohort. Patients who achieved RCB-I had similar survival outcomes as those with pathological complete response (pCR, RCB-0). In whole cohort, for the RCB index and the Miller-Payne system, respectively, C-indexes for DFS were 0.73 and 0.64, for DSS were 0.74 and 0.64. The average RCB score was different among three subtypes (F=9.335, P<0.001). CONCLUSIONS The RCB index and the Miller-Payne system can stratify survival outcome of patients after NAC, and RCB had a superior prediction accuracy, especially for triple-negative breast cancer (TNBC). New cut-off value should be sought in order to improve prediction accuracy.
Collapse
Affiliation(s)
- Wei Wang
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Yinhua Liu
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Hong Zhang
- Pathology Department, Peking University First Hospital, Beijing, China
| | - Shuang Zhang
- Pathology Department, Peking University First Hospital, Beijing, China
| | - Xuening Duan
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Jingming Ye
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Ling Xu
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Jianxin Zhao
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Yuanjia Cheng
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Qian Liu
- Breast Disease Center, Peking University First Hospital, Beijing, China
| |
Collapse
|
31
|
Neoadjuvant hormonal therapy before radical prostatectomy in high-risk prostate cancer. Nat Rev Urol 2021; 18:739-762. [PMID: 34526701 DOI: 10.1038/s41585-021-00514-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 02/08/2023]
Abstract
Patients with high-risk prostate cancer treated with curative intent are at an increased risk of biochemical recurrence, metastatic progression and cancer-related death compared with patients treated for low-risk or intermediate-risk disease. Thus, these patients often need multimodal therapy to achieve complete disease control. Over the past two decades, multiple studies on the use of neoadjuvant treatment have been performed using conventional androgen deprivation therapy, which comprises luteinizing hormone-releasing hormone agonists or antagonists and/or first-line anti-androgens. However, despite results from these studies demonstrating a reduction in positive surgical margins and tumour volume, no benefit has been observed in hard oncological end points, such as cancer-related death. The introduction of potent androgen receptor signalling inhibitors (ARSIs), such as abiraterone, apalutamide, enzalutamide and darolutamide, has led to a renewed interest in using neoadjuvant hormonal treatment in high-risk prostate cancer. The addition of ARSIs to androgen deprivation therapy has demonstrated substantial survival benefits in the metastatic castration-resistant, non-metastatic castration-resistant and metastatic hormone-sensitive settings. Intuitively, a similar survival effect can be expected when applying ARSIs as a neoadjuvant strategy in high-risk prostate cancer. Most studies on neoadjuvant ARSIs use a pathological end point as a surrogate for long-term oncological outcome. However, no consensus yet exists regarding the ideal definition of pathological response following neoadjuvant hormonal therapy and pathologists might encounter difficulties in determining pathological response in hormonally treated prostate specimens. The neoadjuvant setting also provides opportunities to gain insight into resistance mechanisms against neoadjuvant hormonal therapy and, consequently, to guide personalized therapy.
Collapse
|
32
|
Van Bockstal MR, François A, Altinay S, Arnould L, Balkenhol M, Broeckx G, Burguès O, Colpaert C, Dedeurwaerdere F, Dessauvagie B, Duwel V, Floris G, Fox S, Gerosa C, Hastir D, Jaffer S, Kurpershoek E, Lacroix-Triki M, Laka A, Lambein K, MacGrogan GM, Marchio C, Martinez MDM, Nofech-Mozes S, Peeters D, Ravarino A, Reisenbichler E, Resetkova E, Sanati S, Schelfhout AM, Schelfhout V, Shaaban A, Sinke R, Stanciu-Pop CM, van Deurzen CHM, Van de Vijver KK, Van Rompuy AS, Vincent-Salomon A, Wen H, Wong S, Bouzin C, Galant C. Interobserver variability in the assessment of stromal tumor-infiltrating lymphocytes (sTILs) in triple-negative invasive breast carcinoma influences the association with pathological complete response: the IVITA study. Mod Pathol 2021; 34:2130-2140. [PMID: 34218258 PMCID: PMC8595512 DOI: 10.1038/s41379-021-00865-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
High stromal tumor-infiltrating lymphocytes (sTILs) in triple-negative breast cancer (TNBC) are associated with pathological complete response (pCR) after neoadjuvant chemotherapy (NAC). Histopathological assessment of sTILs in TNBC biopsies is characterized by substantial interobserver variability, but it is unknown whether this affects its association with pCR. Here, we aimed to investigate the degree of interobserver variability in an international study, and its impact on the relationship between sTILs and pCR. Forty pathologists assessed sTILs as a percentage in digitalized biopsy slides, originating from 41 TNBC patients who were treated with NAC followed by surgery. Pathological response was quantified by the MD Anderson Residual Cancer Burden (RCB) score. Intraclass correlation coefficients (ICCs) were calculated per pathologist duo and Bland-Altman plots were constructed. The relation between sTILs and pCR or RCB class was investigated. The ICCs ranged from -0.376 to 0.947 (mean: 0.659), indicating substantial interobserver variability. Nevertheless, high sTILs scores were significantly associated with pCR for 36 participants (90%), and with RCB class for eight participants (20%). Post hoc sTILs cutoffs at 20% and 40% resulted in variable associations with pCR. The sTILs in TNBC with RCB-II and RCB-III were intermediate to those of RCB-0 and RCB-I, with lowest sTILs observed in RCB-I. However, the limited number of RCB-I cases precludes any definite conclusions due to lack of power, and this observation therefore requires further investigation. In conclusion, sTILs are a robust marker for pCR at the group level. However, if sTILs are to be used to guide the NAC scheme for individual patients, the observed interobserver variability might substantially affect the chance of obtaining a pCR. Future studies should determine the 'ideal' sTILs threshold, and attempt to fine-tune the patient selection for sTILs-based de-escalation of NAC regimens. At present, there is insufficient evidence for robust and reproducible sTILs-guided therapeutic decisions.
Collapse
Affiliation(s)
- Mieke R. Van Bockstal
- Department of pathology, Cliniques universitaires Saint-Luc Bruxelles, Avenue Hippocrate 10, Woluwé-Saint-Lambert 1200, Belgium
| | - Aline François
- Department of pathology, Cliniques universitaires Saint-Luc Bruxelles, Avenue Hippocrate 10, Woluwé-Saint-Lambert 1200, Belgium
| | - Serdar Altinay
- Department of Pathology, University of Health Sciences, Bakirköy Dr. Sadi Konuk Health Application and Research Center, 34147 Istanbul, Turkey
| | - Laurent Arnould
- Département de Biologie et de Pathologie des Tumeurs, Centre George-François Leclerc, 1 Rue Pr. Marion, 21000 Dijon, France
| | - Maschenka Balkenhol
- Department of Pathology, Radboud University Medical Center, PO Box 9100, 6500, HB Nijmegen, The Netherlands
| | - Glenn Broeckx
- Department of Pathology, University Hospital Antwerp, Drie Eikenstraat 655, 2650 Edegem, Belgium
| | - Octavio Burguès
- Department of Pathology, Hospital Clínico Universitario de Valencia, Av. De Blasco Ibáñez 17, 46010 València, Valencia, Spain
| | - Cecile Colpaert
- Department of Pathology, AZ Turnhout Campus Sint-Jozef, Steenweg op Merksplas 44, 2300 Turnhout, Belgium
| | | | - Benjamin Dessauvagie
- Division of Pathology and Laboratory Medicine, Medical School, The University of Western Australia, Crawley, WA 6009, Australia,Anatomical Pathology, PathWest Laboratory Medicine WA, Perth, Australia
| | - Valérie Duwel
- Department of pathology, AZ Klina Brasschaat, Augustijnslei 100, 2930 Brasschaat, Belgium
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, KU Leuven – University of Leuven, Herestraat 49, 3000 Leuven, Belgium,Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven – University of Leuven, Leuven, Belgium
| | - Stephen Fox
- Department of Pathology, Peter MacCallum Cancer Center and the University of Melbourne, Melbourne, Vic 3000, Australia
| | - Clara Gerosa
- Department of Pathology, University of Cagliari, AOU San Giovanni di Dio, Via Ospedale 54, 09124 Cagliari, Italy
| | - Delfyne Hastir
- Institute of Pathology, Lausanne University Hospital, Rue du Bugnon 25, CH-1011 Lausanne, Switzerland
| | - Shabnam Jaffer
- Department of Pathology, Mount Sinai Hospital and Icahn School of Medicine, New York, New York, NY10029 USA
| | | | - Magali Lacroix-Triki
- Department of Pathology, Gustave-Roussy Cancer Campus, 114 Rue Edouard-Vaillant, 94805 Villejuif, France
| | - Andoni Laka
- Department of Pathology, Clinique Notre-Dame de Grâce (CNDG), Chaussée de Nivelles 212, 6041 Gosselies, Belgium
| | - Kathleen Lambein
- Department of Pathology, AZ St Lucas Hospital, Groenebriel 1, 9000 Ghent, Belgium
| | - Gaëtan Marie MacGrogan
- Surgical Pathology Unit, Department of Pathobiology, Institut Bergonié, F-33076 Bordeaux, France
| | - Caterina Marchio
- Department of Medical Sciences, University of Turin, 10126 Torino, Italy,Pathology Unit, FPO-IRCCS, Candiolo Cancer Institute, Candiolo, Italy
| | | | - Sharon Nofech-Mozes
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Ontario, ON M4N 3M5, Canada
| | - Dieter Peeters
- Department of Pathology, AZ St Maarten, Liersesteenweg 435, 2800 Mechelen, Belgium,Histopathology, Imaging and Quantification Unit, HistoGeneX, Sint-Bavostraat 78, 2610 Antwerp, Belgium
| | - Alberto Ravarino
- Department of Pathology, University of Cagliari, AOU San Giovanni di Dio, Via Ospedale 54, 09124 Cagliari, Italy
| | - Emily Reisenbichler
- Department of Pathology, Yale School of Medicine, Yale New Haven Hospital, 310 Cedar Street, New Haven, CT06510, United States
| | - Erika Resetkova
- The University of Texas MD Anderson Cancer Center, Houston TX77030, Texas, USA
| | - Souzan Sanati
- Department of Pathology and Lab Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Rm8612, Los Angeles, CA90048, United States
| | - Anne-Marie Schelfhout
- Department of Pathology, Onze-Lieve-Vrouwziekenhuis Aalst, Moorselbaan 164, 9300 Aalst, Belgium
| | - Vera Schelfhout
- Department of Pathology, AZ St Maarten, Liersesteenweg 435, 2800 Mechelen, Belgium
| | - Abeer Shaaban
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, University of Birmingham, Birmingham B15 2GW, United Kingdom
| | - Renata Sinke
- Pathan BV, Kleiweg 500, 3045 PM Rotterdam, The Netherlands
| | - Claudia M Stanciu-Pop
- Department of Pathology, CHU UCL Namur, Site Godinne, Avenue Docteur G. Thérasse 1, 5530 Yvoir, Belgium
| | - Carolien HM van Deurzen
- Department of Pathology, Erasmus Medical Center Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Koen K Van de Vijver
- Department of Pathology, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Anne-Sophie Van Rompuy
- Department of Pathology, University Hospitals Leuven, KU Leuven – University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Anne Vincent-Salomon
- Pôle de Médicine Diagnostique & Théranostique, INSERM U934, Institut Curie, 26 Rue d’Ulm, 75248 Paris Cedex 05, France
| | - Hannah Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Serena Wong
- Department of Pathology, Yale School of Medicine, Yale New Haven Hospital, 310 Cedar Street, New Haven, CT06510, United States
| | - Caroline Bouzin
- 2IP IREC Imaging Platform, Institute of Clinical and Experimental Research (IREC), Université catholique de Louvain, Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Christine Galant
- Department of pathology, Cliniques universitaires Saint-Luc Bruxelles, Avenue Hippocrate 10, Woluwé-Saint-Lambert 1200, Belgium,Institute of Clinical and Experimental Research (IREC), Université catholique de Louvain, Avenue Hippocrate 55, 1200 Brussels, Belgium
| |
Collapse
|
33
|
Symmans WF, Yau C, Chen YY, Balassanian R, Klein ME, Pusztai L, Nanda R, Parker BA, Datnow B, Krings G, Wei S, Feldman MD, Duan X, Chen B, Sattar H, Khazai L, Zeck JC, Sams S, Mhawech-Fauceglia P, Rendi M, Sahoo S, Ocal IT, Fan F, LeBeau LG, Vinh T, Troxell ML, Chien AJ, Wallace AM, Forero-Torres A, Ellis E, Albain KS, Murthy RK, Boughey JC, Liu MC, Haley BB, Elias AD, Clark AS, Kemmer K, Isaacs C, Lang JE, Han HS, Edmiston K, Viscusi RK, Northfelt DW, Khan QJ, Leyland-Jones B, Venters SJ, Shad S, Matthews JB, Asare SM, Buxton M, Asare AL, Rugo HS, Schwab RB, Helsten T, Hylton NM, van 't Veer L, Perlmutter J, DeMichele AM, Yee D, Berry DA, Esserman LJ. Assessment of Residual Cancer Burden and Event-Free Survival in Neoadjuvant Treatment for High-risk Breast Cancer: An Analysis of Data From the I-SPY2 Randomized Clinical Trial. JAMA Oncol 2021; 7:1654-1663. [PMID: 34529000 DOI: 10.1001/jamaoncol.2021.3690] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Importance Residual cancer burden (RCB) distributions may improve the interpretation of efficacy in neoadjuvant breast cancer trials. Objective To compare RCB distributions between randomized control and investigational treatments within subtypes of breast cancer and explore the relationship with survival. Design, Setting, and Participants The I-SPY2 is a multicenter, platform adaptive, randomized clinical trial in the US that compares, by subtype, investigational agents in combination with chemotherapy vs chemotherapy alone in adult women with stage 2/3 breast cancer at high risk of early recurrence. Investigational treatments graduated in a prespecified subtype if there was 85% or greater predicted probability of higher rate of pathologic complete response (pCR) in a confirmatory, 300-patient, 1:1 randomized, neoadjuvant trial in that subtype. Evaluation of a secondary end point was reported from the 10 investigational agents tested in the I-SPY2 trial from March 200 through 2016, and analyzed as of September 9, 2020. The analysis plan included modeling of RCB within subtypes defined by hormone receptor (HR) and ERBB2 status and compared control treatments with investigational treatments that graduated and those that did not graduate. Interventions Neoadjuvant paclitaxel plus/minus 1 of several investigational agents for 12 weeks, then 12 weeks of cyclophosphamide/doxorubicin chemotherapy followed by surgery. Main Outcomes and Measures Residual cancer burden (pathological measure of residual disease) and event-free survival (EFS). Results A total of 938 women (mean [SD] age, 49 [11] years; 66 [7%] Asian, 103 [11%] Black, and 750 [80%] White individuals) from the first 10 investigational agents were included, with a median follow-up of 52 months (IQR, 29 months). Event-free survival worsened significantly per unit of RCB in every subtype of breast cancer (HR-positive/ERBB2-negative: hazard ratio [HZR], 1.75; 95% CI, 1.45-2.16; HR-positive/ERBB2-positive: HZR, 1.55; 95% CI, 1.18-2.05; HR-negative/ERBB2-positive: HZR, 2.39; 95% CI, 1.64-3.49; HR-negative/ERBB2-negative: HZR, 1.99; 95% CI, 1.71-2.31). Prognostic information from RCB was similar from treatments that graduated (HZR, 2.00; 95% CI, 1.57-2.55; 254 [27%]), did not graduate (HZR, 1.87; 95% CI, 1.61-2.17; 486 [52%]), or were control (HZR, 1.79; 95% CI, 1.42-2.26; 198 [21%]). Investigational treatments significantly lowered RCB in HR-negative/ERBB2-negative (graduated and nongraduated treatments) and ERBB2-positive subtypes (graduated treatments), with improved EFS (HZR, 0.61; 95% CI, 0.41-0.93) in the exploratory analysis. Conclusions and Relevance In this randomized clinical trial, the prognostic significance of RCB was consistent regardless of subtype and treatment. Effective neoadjuvant treatments shifted the distribution of RCB in addition to increasing pCR rate and appeared to improve EFS. Using a standardized quantitative method to measure response advances the interpretation of efficacy. Trial Registration ClinicalTrials.gov Identifier: NCT01042379.
Collapse
Affiliation(s)
- W Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Christina Yau
- Department of Surgery, University of California, San Francisco
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco
| | - Ron Balassanian
- Department of Pathology, University of California, San Francisco
| | - Molly E Klein
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Lajos Pusztai
- Department of Medicine, Medical Oncology, Yale University, New Haven, Connecticut
| | - Rita Nanda
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Barbara A Parker
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Brian Datnow
- Department of Pathology, University of California, San Diego, La Jolla
| | - Gregor Krings
- Department of Pathology, University of California, San Francisco
| | - Shi Wei
- Department of Anatomic Pathology, University of Alabama at Birmingham
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia
| | - Xiuzhen Duan
- Department of Pathology, Loyola University, Chicago, Illinois
| | - Beiyun Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Husain Sattar
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Laila Khazai
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Jay C Zeck
- Department of Pathology, Georgetown University, Washington, DC
| | - Sharon Sams
- Department of Pathology, University of Colorado Anschutz Medical Center, Aurora
| | | | - Mara Rendi
- Department of Anatomic Pathology, University of Washington, Seattle
| | - Sunati Sahoo
- Department of Pathology, University of Texas Southwestern, Dallas
| | - Idris Tolgay Ocal
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona
| | - Fang Fan
- Department of Pathology, University of Kansas Medical Center, Kansas City
| | | | - Tuyethoa Vinh
- Department of Pathology, Inova Health System, Fairfax, Virginia
| | - Megan L Troxell
- Department of Pathology, Oregon Health and Science University, Portland
| | - A Jo Chien
- Division of Hematology-Oncology, Department of Medicine, University of California, San Francisco
| | - Anne M Wallace
- Department of Surgery, University of California, San Diego, La Jolla
| | - Andres Forero-Torres
- Division of Hematology-Oncology, Department of Medicine, University of Alabama at Birmingham
| | - Erin Ellis
- Medical Oncology, Swedish Cancer Institute, Seattle, Washington
| | - Kathy S Albain
- Division of Hematology-Oncology, Department of Medicine, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Rashmi K Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Minetta C Liu
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Barbara B Haley
- Division of Hematology-Oncology, Department of Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Anthony D Elias
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora
| | - Amy S Clark
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia
| | - Kathleen Kemmer
- Division of Hematology-Oncology, Department of Medicine, Oregon Health & Science University, Portland
| | - Claudine Isaacs
- Division of Hematology-Oncology, Department of Medicine, Georgetown University, Washington, DC
| | - Julie E Lang
- Department of Surgery, University of Southern California, Los Angeles
| | - Hyo S Han
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Kirsten Edmiston
- Department of Surgery, Inova Schar Cancer Institute, Fairfax, Virginia
| | - Rebecca K Viscusi
- Department of Surgery, University of Arizona Health Sciences, Tucson, Arizona
| | - Donald W Northfelt
- Department of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Qamar J Khan
- Division of Oncology, Department of Medicine, University of Kansas, Kansas City
| | | | - Sara J Venters
- Department of Laboratory Medicine, University of California, San Francisco
| | - Sonal Shad
- Department of Surgery, University of California, San Francisco
| | | | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | - Adam L Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | - Hope S Rugo
- Division of Hematology-Oncology, Department of Medicine, University of California, San Francisco
| | - Richard B Schwab
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Teresa Helsten
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Laura van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco
| | | | - Angela M DeMichele
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia
| | - Douglas Yee
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis
| | | | | |
Collapse
|
34
|
Thompson BM, Chala LF, Shimizu C, Mano MS, Filassi JR, Geyer FC, Torres US, de Mello GGN, da Costa Leite C. Pre-treatment MRI tumor features and post-treatment mammographic findings: may they contribute to refining the prediction of pathologic complete response in post-neoadjuvant breast cancer patients with radiologic complete response on MRI? Eur Radiol 2021; 32:1663-1675. [PMID: 34716780 DOI: 10.1007/s00330-021-08290-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/05/2021] [Accepted: 08/20/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Radiologic complete response (rCR) in breast cancer patients after neoadjuvant chemotherapy (NAC) does not necessarily correlate with pathologic complete response (pCR), a marker traditionally associated with better outcomes. We sought to verify if data extracted from two important steps of the imaging workup (tumor features at pre-treatment MRI and post-treatment mammographic findings) might assist in refining the prediction of pCR in post-NAC patients showing rCR. METHODS A total of 115 post-NAC women with rCR on MRI (2010-2016) were retrospectively assessed. Pre-treatment MRI (lesion morphology, size, and distribution) and post-treatment mammographic findings (calcification, asymmetry, mass, architectural distortion) were assessed, as well as clinical and molecular variables. Bivariate and multivariate analyses evaluated correlation between such variables and pCR. Post-NAC mammographic findings and their correlation with ductal in situ carcinoma (DCIS) were evaluated using Pearson's correlation. RESULTS Tumor distribution at pre-treatment MRI was the only significant predictive imaging feature on multivariate analysis, with multicentric lesions having lower odds of pCR (p = 0.035). There was no significant association between tumor size and morphology with pCR. Mammographic residual calcifications were associated with DCIS (p = 0.009). The receptor subtype remained as a significant predictor, with HR-HER2 + and triple-negative status demonstrating higher odds of pCR on multivariate analyses. CONCLUSIONS Multicentric lesions on pre-NAC MRI were associated with a lower chance of pCR in post-NAC rCR patients. The receptor subtype remained a reliable predictor of pCR. Residual mammographic calcifications correlated with higher odds of malignancy, making the correlation between mammography and MRI essential for surgical planning. Key Points • The presence of a multicentric lesion on pre-NAC MRI, even though the patient reaches a radiologic complete response on MRI, is associated with a lower chance of pCR. • Molecular status of the tumor remained the only significant predictor of pathologic complete response in such patients in the present study. • Post-neoadjuvant residual calcifications found on mammography were related to higher odds of residual malignancy, making the correlation between mammography and MRI essential for surgical planning.
Collapse
Affiliation(s)
- Bruna M Thompson
- Institute of Radiology, Clinics Hospital, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Luciano F Chala
- Fleury Group, Rua Cincinato Braga, 282, Bela Vista, São Paulo, SP, 01333-010, Brazil
| | - Carlos Shimizu
- Institute of Radiology, Clinics Hospital, School of Medicine, University of São Paulo, São Paulo, Brazil.,Fleury Group, Rua Cincinato Braga, 282, Bela Vista, São Paulo, SP, 01333-010, Brazil
| | - Max S Mano
- Department of Oncology, Hospital Sírio Libanês, São Paulo, Brazil
| | - José R Filassi
- Department of Gynecology and Obstetrics, Mastology Section, Instituto Do Câncer Do Estado de São Paulo, São Paulo, Brazil
| | - Felipe C Geyer
- Department of Pathology, Instituto Do Câncer Do Estado de São Paulo, São Paulo, Brazil
| | - Ulysses S Torres
- Fleury Group, Rua Cincinato Braga, 282, Bela Vista, São Paulo, SP, 01333-010, Brazil.
| | | | - Cláudia da Costa Leite
- Institute of Radiology, Clinics Hospital, School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
35
|
HER2 Testing Characteristics Can Predict Residual Cancer Burden following Neoadjuvant Chemotherapy in HER2-Positive Breast Cancer. Int J Breast Cancer 2021; 2021:6684629. [PMID: 34123431 PMCID: PMC8166502 DOI: 10.1155/2021/6684629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/09/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives The response to HER2-targeted neoadjuvant chemotherapy (NAC) in HER2-positive (+) breast cancer can be quantified using residual cancer burden (RCB) pathologic evaluation to predict relapse free/overall survival. However, more information is needed to characterize the relationship between patterns of HER2 testing results and response to NAC. We evaluated clinicopathologic characteristics associated with RCB categories in HER2+ patients who underwent HER2-directed NAC. Methods A retrospective chart review was conducted with Stage I-III HER2+ breast cancer cases following NAC and surgical resection. HER2 immunohistochemistry (IHC) staining and fluorescence in situ hybridization (FISH), histologic/clinical characteristics, hormone receptor status, and RCB scores (RCB-0, RCB-I, RCB-II, and RCB-III) were evaluated. Results 64/151 (42.4%) patients with HER2+ disease had pathologic complete response (pCR). Tumors with suboptimal response (RCB-II and RCB-III) were more likely to demonstrate less than 100% HER2 IHC 3+ staining (p < 0.0001), lower HER2 FISH copies (p < 0.0001), and lower HER2/CEP17 ratios (p = 0.0015) compared to RCB-I and RCB-II responses. Estrogen receptor classification using ≥10% versus ≥1% staining showed greater association with higher RCB categories. Conclusions HER2+ characteristics show differing response to therapy despite all being categorized as positive; tumors with less than 100% IHC 3+ staining, lower HER2 FISH copies, and lower HER2/CEP17 ratios resulted in higher RCB scores.
Collapse
|
36
|
Miglietta F, Dieci MV, Griguolo G, Guarneri V. Neoadjuvant approach as a platform for treatment personalization: focus on HER2-positive and triple-negative breast cancer. Cancer Treat Rev 2021; 98:102222. [PMID: 34023642 DOI: 10.1016/j.ctrv.2021.102222] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/06/2021] [Accepted: 05/09/2021] [Indexed: 10/21/2022]
Abstract
The neoadjuvant setting provides unquestionable clinical benefits for high-risk breast cancer (BC) patients, mainly in terms of expansion of locoregional treatment options and prognostic stratification. Additionally, it is also emerging as a strategical tool in the research field. In the present review, by focusing on HER2-positive and triple-negative subtypes, we examined the role of the neoadjuvant setting as a research platform to facilitate and rationalize the placement of escalation strategies, promote the adoption of biomarker-driven approaches for the investigation of de-escalated treatments, and foster the conduction of comprehensive translational analyses, thus ultimately aiming at pursuing treatment personalization. The solid prognostic role of pathologic complete response after neoadjuvant therapy, and its use as a surrogate endpoint to accelerate the drug approval process were discussed. In this context, available data on escalated treatment strategies capable of enhancing pathologic complete response (pCR) rate or improving prognosis of patients with residual disease (RD) after neoadjuvant treatment, were comprehensively reviewed. We also summarized evidence regarding the possibility of obtaining pCR with de-escalated strategies, with particular emphasis on the role of biomarker-driven approaches for patient selection. Pitfalls of the dichotomy of pCR/RD were also deepened, and data on alternative/complementary biomarkers with a possible clinical relevance in this regard were reviewed.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| |
Collapse
|
37
|
Ciga O, Xu T, Nofech-Mozes S, Noy S, Lu FI, Martel AL. Overcoming the limitations of patch-based learning to detect cancer in whole slide images. Sci Rep 2021; 11:8894. [PMID: 33903725 PMCID: PMC8076327 DOI: 10.1038/s41598-021-88494-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/06/2021] [Indexed: 11/09/2022] Open
Abstract
Whole slide images (WSIs) pose unique challenges when training deep learning models. They are very large which makes it necessary to break each image down into smaller patches for analysis, image features have to be extracted at multiple scales in order to capture both detail and context, and extreme class imbalances may exist. Significant progress has been made in the analysis of these images, thanks largely due to the availability of public annotated datasets. We postulate, however, that even if a method scores well on a challenge task, this success may not translate to good performance in a more clinically relevant workflow. Many datasets consist of image patches which may suffer from data curation bias; other datasets are only labelled at the whole slide level and the lack of annotations across an image may mask erroneous local predictions so long as the final decision is correct. In this paper, we outline the differences between patch or slide-level classification versus methods that need to localize or segment cancer accurately across the whole slide, and we experimentally verify that best practices differ in both cases. We apply a binary cancer detection network on post neoadjuvant therapy breast cancer WSIs to find the tumor bed outlining the extent of cancer, a task which requires sensitivity and precision across the whole slide. We extensively study multiple design choices and their effects on the outcome, including architectures and augmentations. We propose a negative data sampling strategy, which drastically reduces the false positive rate (25% of false positives versus 62.5%) and improves each metric pertinent to our problem, with a 53% reduction in the error of tumor extent. Our results indicate classification performances of image patches versus WSIs are inversely related when the same negative data sampling strategy is used. Specifically, injection of negatives into training data for image patch classification degrades the performance, whereas the performance is improved for slide and pixel-level WSI classification tasks. Furthermore, we find applying extensive augmentations helps more in WSI-based tasks compared to patch-level image classification.
Collapse
Affiliation(s)
- Ozan Ciga
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| | - Tony Xu
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, Canada
| | - Sharon Nofech-Mozes
- Department of Laboratory Medicine and Molecular Diagnostics, University of Toronto, Toronto, Canada
- Division of Anatomic Pathology, Sunnybrook Health Science Center, Toronto, ON, Canada
| | - Shawna Noy
- Sunnybrook Research Institute, Toronto, Canada
| | - Fang-I Lu
- Department of Laboratory Medicine and Molecular Diagnostics, University of Toronto, Toronto, Canada
- Division of Anatomic Pathology, Sunnybrook Health Science Center, Toronto, ON, Canada
| | - Anne L Martel
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| |
Collapse
|
38
|
TIM3 expression on TILs is associated with poor response to neoadjuvant chemotherapy in patients with locally advanced triple-negative breast cancer. BMC Cancer 2021; 21:357. [PMID: 33823818 PMCID: PMC8025357 DOI: 10.1186/s12885-021-08054-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/16/2021] [Indexed: 01/10/2023] Open
Abstract
Background The expression of immune checkpoint receptors (ICRs) on tumor-infiltrating lymphocytes (TILs) is associated with better response to immunotherapies via immune checkpoint inhibitors. Therefore, we investigated various ICR expressions on TILs in patients with locally advanced triple-negative breast cancer (TNBC) after neoadjuvant chemotherapy (NAC). Methods Expressions of ICRs were examined immunohistochemically in surgical specimens (n = 61) using monoclonal antibodies for PDL-1, PD-1, TIM-3, LAG-3, and CTLA-4. Positivity was defined as staining > 1% on TILs. Results The median age was 49 (24–76) years. The majority of patients were clinically T3–4 (n = 31, 50.8%) and clinically N1–3 (n = 58, 95.1%) before NAC. Of those, 82% were found to have CTLA-4 positivity, whereas PD1, PDL-1, LAG3, and TIM-3 expressions on TILs were 62.3, 50.9, 26.2, and 68.9%. A high expression of CTLA-4 was found to be associated with a better chemotherapy response (OR = 7.94, 95% CI: 0.9–70.12, p = 0.06), whereas TIM-3 positivity was contrarily associated with a worse chemotherapy response (OR = 0.253, 95% CI: 0.066–0.974, p = 0.047) as measured by the MDACC Residual Cancer Burden Index. At a 47-month follow-up, ypN0 (DFS; HR = 0.31, 95% CI: 0.12–0.83, p = 0.02 and DSS; HR = 0.21, 95% CI: 0.07–0.62, p = 0.005) and CTLA-4 high expression on TILs (DFS; HR = 0.38, 95% CI: 0.17–0.85, p = 0.019 and DSS; HR = 0.34, 95% CI: 0.15–0.78, p = 0.01) were found to be associated with improved survival. Conclusions These findings demonstrate that CTLA-4, PD-1, PDL-1, and TIM-3 were highly expressed in TNBC. Based on these high expression patterns, further studies directed towards combined therapies are warranted in advanced TNBC in future.
Collapse
|
39
|
Mohan SC, Walcott-Sapp S, Lee MK, Srour MK, Kim S, Amersi FF, Giuliano AE, Chung AP. Alterations in Breast Cancer Biomarkers Following Neoadjuvant Therapy. Ann Surg Oncol 2021; 28:5907-5917. [PMID: 33748896 DOI: 10.1245/s10434-021-09814-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/09/2021] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Biomarker changes in patients with residual disease (RD) after neoadjuvant systemic therapy (NAT) have unclear consequences. This study examined the prevalence of biomarker [hormone receptor (HR) and HER2] change and its effect on disease-free survival (DFS) and overall survival (OS). PATIENTS AND METHODS A total of 303 patients treated with NAT from 2008 to 2016 were identified from a prospective database. Biomarker status at diagnosis was determined and retested after NAT in patients with RD. DFS and OS were compared among three groups: no biomarker change, clinically insignificant change in either ER or PR without alteration in HR status, and clinically significant change in at least one biomarker with resultant change in HR or HER2 status. Subgroups with no change and HR change were examined [HR+HER2- no change, triple negative (TN) no change, HR+HER2- to TN, TN to HR+HER2]. RESULTS Overall, 61.4% of patients had RD. Of these, 32.8% had changes in at least one biomarker. At median follow up of 5.48 years, no biomarker change was associated with improved DFS compared with changes in HR or HER2 status (p = 0.043). In addition, no biomarker change (p = 0.005) and clinically insignificant changes in biomarker status (p = 0.019) were associated with improved OS compared with clinically significant changes in HR or HER2 status. Among subgroups, HR+HER2- to TN was associated with worse DFS (p = 0.029) and OS (p = 0.008) compared with HR+HER2- no change. CONCLUSIONS Among those with RD, biomarker status change was common and impacted survival in subgroups of HR+ or TN disease. Retesting biomarkers after NAT has prognostic implications.
Collapse
Affiliation(s)
| | - Sarah Walcott-Sapp
- Marion-Louise Saltzman Women's Center, Einstein Medical Center Philadelphia, Philadelphia, PA, USA
| | - Minna K Lee
- Division of Surgical Oncology, Department of Surgery, Cedars Sinai Medical Center, West Hollywood, CA, USA
| | - Marissa K Srour
- Division of Surgical Oncology, Department of Surgery, Cedars Sinai Medical Center, West Hollywood, CA, USA
| | - Sungjin Kim
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Samuel Oschin Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Farin F Amersi
- Division of Surgical Oncology, Department of Surgery, Cedars Sinai Medical Center, West Hollywood, CA, USA
| | - Armando E Giuliano
- Division of Surgical Oncology, Department of Surgery, Cedars Sinai Medical Center, West Hollywood, CA, USA
| | - Alice P Chung
- Division of Surgical Oncology, Department of Surgery, Cedars Sinai Medical Center, West Hollywood, CA, USA.
| |
Collapse
|
40
|
Determination of breast cancer prognosis after neoadjuvant chemotherapy: comparison of Residual Cancer Burden (RCB) and Neo-Bioscore. Br J Cancer 2021; 124:1421-1427. [PMID: 33558711 PMCID: PMC8039034 DOI: 10.1038/s41416-020-01251-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 12/02/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background To compare RCB (Residual Cancer Burden) and Neo-Bioscore in terms of prognostic performance and see if adding pathological variables improve these scores. Methods We analysed 750 female patients with invasive breast cancer (BC) treated with neoadjuvant chemotherapy (NAC) at Institut Curie between 2002 and 2012. Scores were compared in global population and by BC subtype using Akaike information criterion (AIC), C-Index (concordance index), calibration curves and after adding lymphovascular invasion (LVI) and pre-/post-NAC TILs levels. Results RCB and Neo-Bioscore were significantly associated to disease-free and overall survival in global population and for triple-negative BC. RCB had the lowest AICs in every BC subtype, corresponding to a better prognostic performance. In global population, C-Index values were poor for RCB (0.66; CI [0.61–0.71]) and fair for Neo-Bioscore (0.70; CI [0.65–0.75]). Scores were well calibrated in global population, but RCB yielded better prognostic performances in each BC subtype. Concordance between the two scores was poor. Adding LVI and TILs improved the performance of both scores. Conclusions Although RCB and Neo-Bioscore had similar prognostic performances, RCB showed better performance in BC subtypes, especially in luminal and TNBC. By generating fewer prognostic categories, RCB enables an easier use in everyday clinical practice.
Collapse
|
41
|
Tetzlaff MT, Adhikari C, Lo S, Rawson RV, Amaria RN, Menzies AM, Wilmott JS, Ferguson PM, Ross MI, Spillane AJ, Vu KA, Ma J, Ning J, Haydu LE, Saw RPM, Wargo JA, Tawbi HA, Gershenwald JE, Long GV, Davies MA, Scolyer RA. Histopathological features of complete pathological response predict recurrence-free survival following neoadjuvant targeted therapy for metastatic melanoma. Ann Oncol 2020; 31:1569-1579. [PMID: 32739408 DOI: 10.1016/j.annonc.2020.07.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/02/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Recent clinical trials demonstrated the safety and efficacy of neoadjuvant dabrafenib and trametinib (DT) among patients with surgically resectable clinical stage III BRAFV600E/K mutant melanoma. Although patients achieving a complete pathological response (pCR) exhibited superior recurrence-free survival (RFS) versus those who did not, 30% of pCR patients relapsed. We sought to identify whether histopathological features of the pathological response further delineated risk of relapse. METHODS Surgical resection specimens from DT-treated patients in two phase 2 clinical trials were reviewed. Histopathological features, including relative amounts of viable tumour, necrosis, melanosis, and fibrosis (hyalinized or immature/proliferative) were assessed for associations with patient outcomes. RESULTS Fifty-nine patients underwent surgical resection following neoadjuvant DT. Patients achieving pCR (49%) had longer RFS compared with patients who did not (P = 0.005). Patients whose treated tumour showed any hyalinized fibrosis had longer RFS versus those without (P = 0.014), whereas necrosis (P = 0.012) and/or immature/proliferative fibrosis (P = 0.026) correlated with shorter RFS. Multivariable analyses showed absence of pCR or presence of immature fibrosis independently predicted shorter RFS. Among pCR patients, mature/hyalinized-type fibrosis correlated with improved RFS (P = 0.035). CONCLUSIONS The extent and composition of the pathological response following neoadjuvant DT in BRAFV600E/K mutant melanoma correlates with RFS, including pCR patients. These findings support the need for detailed histological analysis of specimens collected after neoadjuvant therapy.
Collapse
Affiliation(s)
- M T Tetzlaff
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Translational and Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - C Adhikari
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia
| | - S Lo
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia
| | - R V Rawson
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia; Royal Prince Alfred Hospital, Sydney, Australia; New South Wales Health Pathology, Sydney, Australia
| | - R N Amaria
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A M Menzies
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia; Royal North Shore and Mater Hospitals, Sydney, Australia
| | - J S Wilmott
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia
| | - P M Ferguson
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia; Royal Prince Alfred Hospital, Sydney, Australia; New South Wales Health Pathology, Sydney, Australia
| | - M I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A J Spillane
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia; Royal North Shore and Mater Hospitals, Sydney, Australia
| | - K A Vu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Ma
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L E Haydu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - R P M Saw
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia; Royal Prince Alfred Hospital, Sydney, Australia
| | - J A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - H A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - G V Long
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia; Royal North Shore and Mater Hospitals, Sydney, Australia
| | - M A Davies
- Department of Translational and Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - R A Scolyer
- Melanoma Institute of Australia, The University of Sydney, Sydney, Australia; Royal Prince Alfred Hospital, Sydney, Australia; New South Wales Health Pathology, Sydney, Australia.
| |
Collapse
|
42
|
Han R, Regpala S, Slodkowska E, Nofech-Mozes S, Hanna W, Parra-Herran C, Lu FI. Lack of Standardization in the Processing and Reporting of Post-Neoadjuvant Breast Cancer Specimens. Arch Pathol Lab Med 2020; 144:1262-1270. [PMID: 32142368 DOI: 10.5858/arpa.2019-0539-oa] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The use of neoadjuvant therapy in the management of early-stage invasive breast cancer is increasing. Residual Cancer Burden and other similar tools use pathologic characteristics of post-neoadjuvant therapy breast tumors to determine long-term outcome. However, there are no standardized guidelines for the pathologic evaluation of these specimens in the routine clinical setting. OBJECTIVE.— To assess current practices among Canadian pathologists and pathology assistants with regard to the processing and reporting of post-neoadjuvant therapy breast specimens. DESIGN.— An electronic survey was distributed to pathologists and pathology assistants across Canada. RESULTS.— Sixty-three responses were obtained. A total of 48% (15 of 31) of surveyed pathologists reported familiarity with the Residual Cancer Burden tool. A total of 40% (25 of 63) of respondents reported a lack of routine use of specimen photography, and 35% (22 of 63) reported a lack of routine use of grossing diagrams. There was significant variation with respect to tumor bed sampling; the most common method was to submit 1 block per centimeter of tumor (20 of 63; 32%). There was also significant variation in the method of measuring residual tumor; the most common method was to measure the largest cross-section of residual tumor (16 of 32; 50%). CONCLUSIONS.— There is a need for standardization of the evaluation of post-neoadjuvant therapy breast specimens in the routine clinical setting in Canada. We recommend the routine use of specimen mapping, submitting the largest cross section of tumor bed in toto, reporting tumor size as per American Joint Committee on Cancer and Residual Cancer Burden guidelines, and routinely including measurements of residual tumor cellularity and in situ disease in the final pathology report as per Residual Cancer Burden guidelines.
Collapse
Affiliation(s)
- Rachel Han
- From the Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (Han)
| | - Steffi Regpala
- The Department of Anatomic Pathology, University of Ottawa and Eastern Ontario Regional Laboratory Association, Ottawa, Canada (Regpala)
| | - Elzbieta Slodkowska
- The Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (Slodkowska, Nofech-Mozes, Hanna, Parra-Herran, Lu)
| | - Sharon Nofech-Mozes
- The Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (Slodkowska, Nofech-Mozes, Hanna, Parra-Herran, Lu)
| | - Wedad Hanna
- The Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (Slodkowska, Nofech-Mozes, Hanna, Parra-Herran, Lu)
| | - Carlos Parra-Herran
- The Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (Slodkowska, Nofech-Mozes, Hanna, Parra-Herran, Lu)
| | - Fang-I Lu
- The Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (Slodkowska, Nofech-Mozes, Hanna, Parra-Herran, Lu)
| |
Collapse
|
43
|
da Silva JL, de Paula BHR, Small IA, Thuler LCS, de Melo AC. Sociodemographic, Clinical, and Pathological Factors Influencing Outcomes in Locally Advanced Triple Negative Breast Cancer: A Brazilian Cohort. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2020; 14:1178223420962488. [PMID: 33029072 PMCID: PMC7522837 DOI: 10.1177/1178223420962488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022]
Abstract
Objective: To evaluate the association of sociodemographic, clinical, and pathological factors with response and survival in triple negative breast cancer (TNBC) undergoing neoadjuvant chemotherapy (NACT). Methods: Clinical-pathological and sociodemographic data were obtained from medical records of 235 eligible women with TNBC diagnosed between 2010 and 2014 undergoing NACT and surgery at the Brazilian National Cancer Institute. They have been assessed for pathological complete response (pCR), event-free survival (EFS), and overall survival (OS). Both univariate and multivariate Cox regression analyses were performed. Results: The median follow-up was 64.3 months. Most patients had advanced clinical stage (III: 85.1%; cT3/T4: 86.4%; cN1-3: 74.4%) and high-grade tumors (72.1%). Clinical staging (III vs II, adjusted hazard ratio [HR] = 2.95, P = .012) significantly influenced the pCR rate. Alcohol intake negatively influenced EFS (adjusted HR = 1.67, P = .006) and OS (adjusted HR = 1.89, P = .005). Women with pCR showed better EFS (crude HR = 0.15, P < .001) and OS (crude HR = 0.12, P < .001) compared with non-pCR. The ypT (<0.001) and ypN (<0.001) gradually influenced survival outcomes. Conclusion: Clinical stage III were associated with lower response rate and worse survival. Alcohol intake, pCR, and burden of post-NACT residual disease have shown considerable influence on survival outcomes.
Collapse
Affiliation(s)
- Jesse Lopes da Silva
- Clinical Research Division, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | | | - Isabele Avila Small
- Clinical Research Division, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | | | | |
Collapse
|
44
|
Blanco Sánchez A, Yébenes L, Berjón A, Hardisson D. [Evaluation of pathological response to neoadjuvant chemotherapy in breast cancer: correlation with molecular phenotype]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2020; 54:8-16. [PMID: 33455698 DOI: 10.1016/j.patol.2020.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/23/2020] [Accepted: 07/20/2020] [Indexed: 11/09/2022]
Abstract
INTRODUCTION AND OBJECTIVE Breast cancer can be classified into different molecular subtypes with important therapeutic and prognostic implications. Neoadjuvant chemotherapy (NAC) increases the possibility of performing conservative surgery and allows in vivo testing of the sensitivity of the tumor. Our aim was to evaluate the pathological response to NAC in relation to the molecular phenotype and the different definitions of the pathological response. PATIENTS 228 patients treated with NAC and subsequent surgery between 2012 and 2018 were selected from our breast cancer database. Molecular phenotypes were established based on the criteria of the St Gallen 2013 Conference. Pathological response was evaluated following Miller-Payne (breast) and Sataloff (axilla) classification systems. RESULTS The most frequent molecular phenotype was luminal B/HER2 negative (30.3%), followed by luminal B/HER2 positive (26.3%), triple negative (24.6%), HER2 positive (13.2%), and luminal A (5.7%). The rate of pathological complete response (pCR) was 35.5% in breast and 15.3% in axilla. The rate of pCR considering breast and axilla together was 26.8%. The molecular phenotype with the highest rate of pCR was HER2 positive (66.7%) followed by triple negative (30.4%), luminal B/HER2 positive (21.7%), luminal B/HER2 negative (14.5%), and luminal A (7.7%) (p < 0.001). The same results were found with the different definitions of pCR we evaluated. CONCLUSIONS Complete pathological response to NAC in breast cancer depends largely on the molecular phenotype of the tumor, regardless of the definition of pCR, with the highest response rates in the breast and axilla in the HER2 positive and triple negative phenotypes.
Collapse
Affiliation(s)
| | - Laura Yébenes
- Servicio de Anatomía Patológica, Hospital Universitario La Paz, Madrid, España; Laboratorio de Patología Molecular del Cáncer y Dianas Terapéuticas, Instituto de Investigación del Hospital Universitario La Paz (IdiPAZ), Madrid, España
| | - Alberto Berjón
- Servicio de Anatomía Patológica, Hospital Universitario La Paz, Madrid, España; Laboratorio de Patología Molecular del Cáncer y Dianas Terapéuticas, Instituto de Investigación del Hospital Universitario La Paz (IdiPAZ), Madrid, España
| | - David Hardisson
- Facultad de Medicina. Universidad Autónoma de Madrid, Madrid, España; Servicio de Anatomía Patológica, Hospital Universitario La Paz, Madrid, España; Laboratorio de Patología Molecular del Cáncer y Dianas Terapéuticas, Instituto de Investigación del Hospital Universitario La Paz (IdiPAZ), Madrid, España; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, España.
| |
Collapse
|
45
|
Hamy AS, Darrigues L, Laas E, De Croze D, Topciu L, Lam GT, Evrevin C, Rozette S, Laot L, Lerebours F, Pierga JY, Osdoit M, Faron M, Feron JG, Laé M, Reyal F. Prognostic value of the Residual Cancer Burden index according to breast cancer subtype: Validation on a cohort of BC patients treated by neoadjuvant chemotherapy. PLoS One 2020; 15:e0234191. [PMID: 32579551 PMCID: PMC7313974 DOI: 10.1371/journal.pone.0234191] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/20/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction The Residual Cancer Burden (RCB) quantifies residual disease after neoadjuvant chemotherapy (NAC). Its predictive value has not been validated on large cohorts with long-term follow up. The objective of this work is to independently evaluate the prognostic value of the RCB index depending on BC subtypes (Luminal, HER2-positive and triple negative (TNBCs)). Methods We retrospectively evaluated the RCB index on surgical specimens from a cohort of T1-T3NxM0 BC patients treated with NAC between 2002 and 2012. We analyzed the association between RCB index and relapse-free survival (RFS), overall survival (OS) among the global population, after stratification by BC subtypes. Results 717 patients were included (luminal BC (n = 222, 31%), TNBC (n = 319, 44.5%), HER2-positive (n = 176, 24.5%)). After a median follow-up of 99.9 months, RCB index was significantly associated with RFS. The RCB-0 patients displayed similar prognosis when compared to the RCB-I group, while patients from the RCB-II and RCB-III classes were at increased risk of relapse (RCB-II versus RCB-0: HR = 3.25 CI [2.1–5.1] p<0.001; RCB-III versus RCB-0: HR = 5.6 CI [3.5–8.9] p<0.001). The prognostic impact of RCB index was significant for TNBC and HER2-positive cancers; but not for luminal cancers (Pinteraction = 0.07). The prognosis of RCB-III patients was poor (8-years RFS: 52.7%, 95% CI [44.8–62.0]) particularly in the TNBC subgroup, where the median RFS was 12.7 months. Conclusion RCB index is a reliable prognostic score. RCB accurately identifies patients at a high risk of recurrence (RCB-III) with TNBC or HER2-positive BC who must be offered second-line adjuvant therapies.
Collapse
Affiliation(s)
- Anne-Sophie Hamy
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, INSERM, U932 Immunity and Cancer, Institut Curie, Paris, France
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Lauren Darrigues
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
| | - Enora Laas
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
| | - Diane De Croze
- Department of Tumor Biology, Institut Curie, Saint-Cloud, France
| | - Lucian Topciu
- Department of Tumor Biology, Institut Curie, Paris, France
| | - Giang-Thanh Lam
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
- Department of Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland
| | - Clemence Evrevin
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Sonia Rozette
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Lucie Laot
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
| | | | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
| | - Marie Osdoit
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
| | - Matthieu Faron
- Department of biostatistics and epidemiology, INSERM 1018 CESP Oncostat team, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jean-Guillaume Feron
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
| | - Marick Laé
- Department of Tumor Biology, Institut Curie, Paris, France
| | - Fabien Reyal
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
- Department of Surgery, Faculté de Médecine Descartes, Université Paris, Institut Curie, Paris, France
- * E-mail:
| |
Collapse
|
46
|
von der Lippe Gythfeldt H, Lien T, Tekpli X, Silwal-Pandit L, Borgen E, Garred Ø, Skjerven H, Schlichting E, Lundgren S, Wist E, Naume B, Kristensen V, Børresen-Dale AL, Lingjaerde OC, Engebraaten O. Immune phenotype of tumor microenvironment predicts response to bevacizumab in neoadjuvant treatment of ER-positive breast cancer. Int J Cancer 2020; 147:2515-2525. [PMID: 32488909 DOI: 10.1002/ijc.33108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/02/2020] [Accepted: 04/30/2020] [Indexed: 12/24/2022]
Abstract
Antiangiogenic drugs are potentially a useful supplement to neoadjuvant chemotherapy for a subgroup of patients with human epidermal growth factor receptor 2 (HER2) negative breast cancer, but reliable biomarkers for improved response are lacking. Here, we report on a randomized phase II clinical trial to study the added effect of bevacizumab in neoadjuvant chemotherapy with FEC100 (5-fluorouracil, epirubicin and cyclophosphamide) and taxanes (n = 132 patients). Gene expression from the tumors was obtained before neoadjuvant treatment, and treatment response was evaluated by residual cancer burden (RCB) at time of surgery. Bevacizumab increased the proportion of complete responders (RCB class 0) from 5% to 20% among patients with estrogen receptor (ER) positive tumors (P = .02). Treatment with bevacizumab was associated with improved 8-year disease-free survival (P = .03) among the good responders (RCB class 0 or I). Patients treated with paclitaxel (n = 45) responded better than those treated with docetaxel (n = 21; P = .03). Improved treatment response was associated with higher proliferation rate and an immune phenotype characterized by high presence of classically activated M1 macrophages, activated NK cells and memory activated CD4 T cells. Treatment with bevacizumab increased the number of adverse events, including hemorrhage, hypertension, infection and febrile neutropenia, but despite this, the ECOG status was not affected.
Collapse
Affiliation(s)
- Hedda von der Lippe Gythfeldt
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Tonje Lien
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Xavier Tekpli
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Laxmi Silwal-Pandit
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Helle Skjerven
- Department of Breast and Endocrine Surgery, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Ellen Schlichting
- Department of Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway
| | - Steinar Lundgren
- Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Erik Wist
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Naume
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vessela Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Christian Lingjaerde
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Informatics, University of Oslo, Oslo, Norway.,KG Jebsen Centre for B-Cell Malignancies, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav Engebraaten
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
47
|
Bossuyt V, Spring L. Pathologic evaluation of response to neoadjuvant therapy drives treatment changes and improves long-term outcomes for breast cancer patients. Breast J 2020; 26:1189-1198. [PMID: 32468652 DOI: 10.1111/tbj.13864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/03/2020] [Indexed: 11/29/2022]
Abstract
Systemic therapy for breast cancer may be given before (neoadjuvant) or after (adjuvant) surgery. When neoadjuvant systemic therapy is given, response to treatment can be evaluated. However, some prognostic information (for example, pathologic tumor size pretreatment) is then lost and pathologic evaluation of breast specimens after neoadjuvant therapy is more difficult. Pathologic complete response (pCR), defined as no invasive disease in the breast (ypT0/is or ypT0) and no disease in all sampled lymph nodes (ypN0), identifies patients with a lower risk of recurrence or death compared to those with residual disease. Multidisciplinary collaboration, marking of the tumor site and any lymph node involvement pretreatment, and access to specimen imaging to facilitate correlation of gross and microscopic findings are critical for accurate determination of pCR. For HER2-positive and triple negative tumors requiring systemic therapy, giving the treatment before surgery identifies a high-risk group of patients that can receive additional adjuvant therapy after surgery if a pCR is not achieved. Recent clinical trials have demonstrated that this approach reduced recurrence risk. More than ever, pathologic evaluation of response to neoadjuvant systemic therapy directs treatment received after surgery. Using a single standardized protocol for sampling of the post-neoadjuvant surgical specimen allows pathologists to ensure accurate determination of pCR or residual disease and quantify residual disease. Residual cancer burden (RCB) and AJCC stage provide complementary quantitative information about residual disease and prognosis.
Collapse
Affiliation(s)
- Veerle Bossuyt
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Laura Spring
- Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Arispe Angulo KR, Jawa Z, Visotcky A, Majidi SS, Chitambar CR, Jorns JM. A high mitotic score in breast cancer after neoadjuvant chemotherapy is predictive of outcome and associated with a distinct morphology. Histopathology 2020; 76:661-670. [PMID: 31849088 DOI: 10.1111/his.14049] [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: 10/01/2019] [Revised: 11/14/2019] [Accepted: 12/13/2019] [Indexed: 11/28/2022]
Abstract
AIMS Neoadjuvant chemotherapy (NAC) is frequently used for the treatment of breast cancer. We sought to analyse the clinical, morphological and immunohistochemical features of tumours from patients who did not achieve pathological complete response following NAC. METHODS AND RESULTS We identified stage I-III post-NAC breast cancers from surgical resections (2000-2016) with evaluable residual invasive carcinoma [ypT1a(m) or greater and ≥15% tumour cellularity]. One hundred and forty-three tumours from 142 patients were included. On univariable analysis, a high (score 3) post-NAC mitotic score (as compared with 1 or 2) was significantly associated with invasive ductal carcinoma (IDC) subtype (P = 0.023), high grade, pushing borders with zones of necrosis, hormone receptor and triple-negative status, lack of hormonal therapy, higher cellularity (P < 0.001), and a higher percentage of tumour-infiltrating lymphocytes (P = 0.016). Multivariable analysis showed a high post-NAC mitotic score to be significantly associated with recurrence, distant metastasis, and shortened survival (hazard ratios of 5.73, 4.49, and 3.68, respectively). High post-NAC mitotic score tumours (n = 32) were IDC and had a high Ki67 proliferation index (median, 55%). Of these, 24 (75%) had pushing borders with zones of necrosis; 19 (79.2%) of these had necrosis on preoperative imaging, and 24 (75%), 15 (46.9%) and four (12.5%) lacked androgen receptor, GATA-3 and cytokeratin 18 expression, respectively. CONCLUSIONS High post-NAC mitotic score breast cancers cause high morbidity and mortality, frequently have pushing borders and zones of necrosis, and may show loss of common 'breast cancer markers'. Our findings support that necrosis in pretreatment studies and post-NAC mitotic score should be routinely reported, as they offer significant additional prognostic information to guide management.
Collapse
Affiliation(s)
| | | | - Alexis Visotcky
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shadie S Majidi
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher R Chitambar
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Julie M Jorns
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
49
|
Travis WD, Dacic S, Wistuba I, Sholl L, Adusumilli P, Bubendorf L, Bunn P, Cascone T, Chaft J, Chen G, Chou TY, Cooper W, Erasmus JJ, Ferreira CG, Goo JM, Heymach J, Hirsch FR, Horinouchi H, Kerr K, Kris M, Jain D, Kim YT, Lopez-Rios F, Lu S, Mitsudomi T, Moreira A, Motoi N, Nicholson AG, Oliveira R, Papotti M, Pastorino U, Paz-Ares L, Pelosi G, Poleri C, Provencio M, Roden AC, Scagliotti G, Swisher SG, Thunnissen E, Tsao MS, Vansteenkiste J, Weder W, Yatabe Y. IASLC Multidisciplinary Recommendations for Pathologic Assessment of Lung Cancer Resection Specimens After Neoadjuvant Therapy. J Thorac Oncol 2020; 15:709-740. [PMID: 32004713 DOI: 10.1016/j.jtho.2020.01.005] [Citation(s) in RCA: 266] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/25/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022]
Abstract
Currently, there is no established guidance on how to process and evaluate resected lung cancer specimens after neoadjuvant therapy in the setting of clinical trials and clinical practice. There is also a lack of precise definitions on the degree of pathologic response, including major pathologic response or complete pathologic response. For other cancers such as osteosarcoma and colorectal, breast, and esophageal carcinomas, there have been multiple studies investigating pathologic assessment of the effects of neoadjuvant therapy, including some detailed recommendations on how to handle these specimens. A comprehensive mapping approach to gross and histologic processing of osteosarcomas after induction therapy has been used for over 40 years. The purpose of this article is to outline detailed recommendations on how to process lung cancer resection specimens and to define pathologic response, including major pathologic response or complete pathologic response after neoadjuvant therapy. A standardized approach is recommended to assess the percentages of (1) viable tumor, (2) necrosis, and (3) stroma (including inflammation and fibrosis) with a total adding up to 100%. This is recommended for all systemic therapies, including chemotherapy, chemoradiation, molecular-targeted therapy, immunotherapy, or any future novel therapies yet to be discovered, whether administered alone or in combination. Specific issues may differ for certain therapies such as immunotherapy, but the grossing process should be similar, and the histologic evaluation should contain these basic elements. Standard pathologic response assessment should allow for comparisons between different therapies and correlations with disease-free survival and overall survival in ongoing and future trials. The International Association for the Study of Lung Cancer has an effort to collect such data from existing and future clinical trials. These recommendations are intended as guidance for clinical trials, although it is hoped they can be viewed as suggestion for good clinical practice outside of clinical trials, to improve consistency of pathologic assessment of treatment response.
Collapse
Affiliation(s)
- William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ignacio Wistuba
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Prasad Adusumilli
- Thoracic Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas Bubendorf
- Department of Pathology, University of Basel, Basel, Switzerland
| | - Paul Bunn
- Medical Oncology, Colorado University School of Medicine, Aurora, Colorado
| | - Tina Cascone
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Jamie Chaft
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gang Chen
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | | | - Wendy Cooper
- Department of Pathology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Jeremy J Erasmus
- Department of Radiology, MD Anderson Cancer Center, Houston, Texas
| | | | - Jin-Mo Goo
- Department of Radiology, Seoul National University College of Medicine, Seoul, South Korea
| | - John Heymach
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute at Mount Sinai, New York, New York
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Keith Kerr
- Department of Pathology, Aberdeen University Medical School, Aberdeen, Scotland
| | - Mark Kris
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Young T Kim
- Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Fernando Lopez-Rios
- Laboratorio de Dianas Terapeuticas, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai, China
| | - Tetsuya Mitsudomi
- Thoracic Surgery, Kinki University Faculty of Medicine, Osaka-Sayama, Japan
| | - Andre Moreira
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Noriko Motoi
- Department of Pathology, Mational Cancer Center, Tokyo, Japan
| | - Andrew G Nicholson
- Department of Pathology, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | | | - Mauro Papotti
- Department of Pathology, University of Turin, Torino, Italy
| | - Ugo Pastorino
- Thoracic Surgery Division, Istituto Nazionale Tumor, Milan, Italy
| | - Luis Paz-Ares
- Medical Oncology, National Oncology Research Center, Madrid, Spain
| | | | - Claudia Poleri
- Office of Pathology Consultants, Buenos Aries, Argentina
| | - Mariano Provencio
- Oncology Department, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Anja C Roden
- Department of Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Yasushi Yatabe
- Department of Pathology, Mational Cancer Center, Tokyo, Japan
| |
Collapse
|
50
|
Barrow-McGee R, Procter J, Owen J, Woodman N, Lombardelli C, Kothari A, Kovacs T, Douek M, George S, Barry PA, Ramsey K, Gibson A, Buus R, Holgersen E, Natrajan R, Haider S, Shattock MJ, Gillett C, Tutt AN, Pinder SE, Naidoo K. Real-time ex vivo perfusion of human lymph nodes invaded by cancer (REPLICANT): a feasibility study. J Pathol 2019; 250:262-274. [PMID: 31755096 PMCID: PMC7065097 DOI: 10.1002/path.5367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/06/2019] [Accepted: 11/18/2019] [Indexed: 01/18/2023]
Abstract
Understanding how breast cancer (BC) grows in axillary lymph nodes (ALNs), and refining how therapies might halt that process, is clinically important. However, modelling the complex ALN microenvironment is difficult, and no human models exist at present. We harvested ALNs from ten BC patients, and perfused them at 37 °C ex vivo for up to 24 h. Controlled autologous testing showed that ALNs remain viable after 24 h of ex vivo perfusion: haematoxylin and eosin-stained histological appearance and proliferation (by Ki67 immunohistochemistry) did not change significantly over time for any perfused ALN compared with a control from time-point zero. Furthermore, targeted gene expression analysis (NanoString PanCancer IO360 panel) showed that only 21/750 genes were differentially expressed between control and perfused ALNs (|log2 FC| > 1 and q < 0.1): none were involved in apoptosis and metabolism, but rather all 21 genes were involved in immune function and angiogenesis. During perfusion, tissue acid-base balance remained stable. Interestingly, the flow rate increased (p < 0.001) in cancer-replaced (i.e. metastasis occupied more than 90% of the surface area on multiple levels) compared to cancer-free nodes (i.e. nodes with no metastasis on multiple sections). CXCL11 transcripts were significantly more abundant in cancer-replaced nodes, while CXCL12 transcripts were significantly more abundant in cancer-free nodes. These cytokines were also detected in the circulating perfusate. Monoclonal antibodies (nivolumab and trastuzumab) were administered into a further three ALNs to confirm perfusion efficacy. These drugs saturated the nodes; nivolumab even induced cancer cell death. Normothermic ALN perfusion is not only feasible but sustains the tumour microenvironment ex vivo for scientific investigation. This model could facilitate the identification of actionable immuno-oncology targets. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Rachel Barrow-McGee
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Julia Procter
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Julie Owen
- King's Health Partners Cancer Biobank, Guy's Comprehensive Cancer Centre, London, UK
| | - Natalie Woodman
- King's Health Partners Cancer Biobank, Guy's Comprehensive Cancer Centre, London, UK
| | - Cristina Lombardelli
- King's Health Partners Cancer Biobank, Guy's Comprehensive Cancer Centre, London, UK
| | | | - Tibor Kovacs
- Guy's and St. Thomas' Hospital NHS Foundation Trust, London, UK
| | - Michael Douek
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Comprehensive Cancer Centre, London, UK
| | - Simi George
- Department of Cellular Pathology, Guy's and St. Thomas' Hospital NHS Foundation Trust, London, UK
| | | | | | - Amy Gibson
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Richard Buus
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK.,Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Erle Holgersen
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Rachael Natrajan
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Syed Haider
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Michael J Shattock
- British Heart Foundation Centre of Research Excellence, King's College London, St Thomas' Hospital, London, UK
| | - Cheryl Gillett
- King's Health Partners Cancer Biobank, Guy's Comprehensive Cancer Centre, London, UK
| | - Andrew Nj Tutt
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Sarah E Pinder
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Comprehensive Cancer Centre, London, UK
| | - Kalnisha Naidoo
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK.,Department of Cellular Pathology, Guy's and St. Thomas' Hospital NHS Foundation Trust, London, UK
| |
Collapse
|