1
|
Guerrero-Zotano Á, Pérez-García JM, Ruiz-Borrego M, Bermejo B, Gil-Gil M, de la Haba J, Alba Conejo E, Quiroga V, Carañana V, Urruticoechea A, Morales S, Bellet M, Antón A, Fernández-Abad M, Sánchez-Rovira P, Calabuig L, Pérez-Escuredo J, Sampayo-Cordero M, Cortés J, Llombart-Cussac A. Neoadjuvant letrozole and palbociclib in patients with HR-positive/HER2-negative early breast cancer and Oncotype DX Recurrence Score ≥18: DxCARTES study. ESMO Open 2024; 9:103733. [PMID: 39353215 PMCID: PMC11472226 DOI: 10.1016/j.esmoop.2024.103733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/25/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The effect of the addition of cyclin-dependent kinases 4 and 6 inhibitors to endocrine therapy in terms of molecular downstaging remains undetermined. Switching from a high-risk to a low risk Recurrence Score (RS) group could provide useful information to identify patients who might not require chemotherapy. The purpose of this study was to assess the biological and clinical activity of letrozole plus palbociclib as neoadjuvant treatment for patients with hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative early breast cancer with an initial Oncotype DX RS ≥18. PATIENTS AND METHODS Participants were women aged ≥18 years with HR-positive/HER2-negative, Ki67 ≥ 20%, stage II-IIIB early breast cancer with a baseline RS ≥18. Eligible patients with a pretreatment RS 18-25 (cohort A) and 26-100 (cohort B) received six 28-day cycles of letrozole (2.5 mg per day; plus goserelin if pre- or perimenopausal) plus palbociclib (125 mg per day; 3/1 schedule) before surgery. The primary endpoint for both cohorts was the proportion of patients who achieved an RS ≤25 at surgery or a pathological complete response (pCR). RESULTS A total of 67 patients were enrolled, among which 65 were assessable for the primary endpoint (32 patients in cohort A and 33 in cohort B). At surgery, 22 (68.8%) patients in cohort A and 18 (54.5%) patients in cohort B had an RS ≤25 or a pCR [only 1 (3.0%) patient in cohort B], meeting the primary endpoint in cohort B (P < 0.01), but not in cohort A (P = 0.98). No new safety signals were identified. CONCLUSIONS The efficacy of neoadjuvant treatment with letrozole plus palbociclib does not seem to depend on pretreatment RS for patients with RS ≥18. However, around half of patients with HR-positive/HER2-negative early breast cancer with an RS 26-100 at baseline achieved molecular downstaging with this regimen.
Collapse
Affiliation(s)
| | - J M Pérez-García
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - M Ruiz-Borrego
- Hospital For Universitario Virgen del Rocío, Sevilla, Spain
| | - B Bermejo
- Hospital Clínico Universitario de Valencia, Valencia, Spain; Biomedical Research Institute INCLIVA, Valencia, Spain
| | - M Gil-Gil
- Institut Català d'Oncologia L'Hospitalet, Hospitalet de Llobregat, Barcelona, Spain
| | - J de la Haba
- Hospital Universitario Reina Sofía, Cordoba, Spain
| | - E Alba Conejo
- Unidad de Gestión Clínica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, Malaga, Spain; Faculty of Medicine, The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), University of Málaga, Malaga, Spain; Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), Madrid, Spain
| | - V Quiroga
- Hospital Clínico Universitario Virgen de la Victoria; Instituto de Investigación Biomédica de Málaga, IBIMA; GEICAM, Spanish Breast Cancer Group, Málaga, Spain
| | - V Carañana
- Hospital Arnau de Vilanova de Valencia, Valencia, Spain
| | - A Urruticoechea
- Gipuzkoa Cancer Unit - BioGipuzkoa, Osakidetza, Donostia, Spain
| | - S Morales
- Hospital Arnau de Vilanova de Lleida, Lleida, Spain
| | - M Bellet
- Hospital Universitari Vall d'Hebrón, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - A Antón
- Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | | | - L Calabuig
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - J Pérez-Escuredo
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - M Sampayo-Cordero
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - J Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil); Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain; IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - A Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil); Hospital Arnau de Vilanova, Valencia, Spain; Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain.
| |
Collapse
|
2
|
Li X, Ruan Z, Yang S, Yang Q, Li J, Hu M. Bioinformatic-Experimental Screening Uncovers Multiple Targets for Increase of MHC-I Expression through Activating the Interferon Response in Breast Cancer. Int J Mol Sci 2024; 25:10546. [PMID: 39408874 PMCID: PMC11476581 DOI: 10.3390/ijms251910546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Expression of major histocompatibility complex I (MHC-I) on tumor cells is extremely important for the antitumor immune response for its essential role in activating various immune cells, including tumor-specific CD8+ T cells. Cancers of lower MHC-I expression commonly exhibit less immune cell infiltration and worse prognosis in clinic. In this study, we conducted bioinformatic-experimental screening to identify potential gene targets to enhance MHC-I expression in breast cancer (BRCA). Through a combination of MHC-I scoring, gene expression correlation analysis, survival prognostication, and Cibersort tumor-infiltrated lymphocytes (TILs) scoring, we identify 144 genes negatively correlated with both MHC-I expression and TILs in breast cancer. Furthermore, we verified partially according to KEGG functional enrichment or gene-dependency analysis and figured out multiple genes, including PIP5K1A, NCKAP1, CYFIP1, DIS3, TBP, and EXOC1, as effective gene targets for increasing MHC-I expression in breast cancer. Mechanistically, knockout of each of these genes activated the intrinsic interferon response in breast cancer cells, which not only promoted MHC-I expression but also caused immunogenic cell death of breast cancer. Finally, the scRNA-seq confirmed the negative correlation of PIP5K1A et al. with TILs in breast cancer patients. Collectively, we identified multiple gene targets for an increase in MHC-I expression in breast cancer in this study.
Collapse
Affiliation(s)
| | | | | | | | - Jinpeng Li
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, State Key Laboratory of Virology, Wuhan University, Wuhan 430072, China; (X.L.); (Z.R.); (S.Y.); (Q.Y.)
| | - Mingming Hu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, State Key Laboratory of Virology, Wuhan University, Wuhan 430072, China; (X.L.); (Z.R.); (S.Y.); (Q.Y.)
| |
Collapse
|
3
|
Zheng G, Hou J, Shu Z, Peng J, Han L, Yuan Z, He X, Gong X. Prediction of neoadjuvant chemotherapy pathological complete response for breast cancer based on radiomics nomogram of intratumoral and derived tissue. BMC Med Imaging 2024; 24:22. [PMID: 38245712 PMCID: PMC10800060 DOI: 10.1186/s12880-024-01198-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Non-invasive identification of breast cancer (BCa) patients with pathological complete response (pCR) after neoadjuvant chemotherapy (NACT) is critical to determine appropriate surgical strategies and guide the resection range of tumor. This study aimed to examine the effectiveness of a nomogram created by combining radiomics signatures from both intratumoral and derived tissues with clinical characteristics for predicting pCR after NACT. METHODS The clinical data of 133 BCa patients were analyzed retrospectively and divided into training and validation sets. The radiomics features for Intratumoral, peritumoral, and background parenchymal enhancement (BPE) in the training set were dimensionalized. Logistic regression analysis was used to select the optimal feature set, and a radiomics signature was constructed using a decision tree. The signature was combined with clinical features to build joint models and generate nomograms. The area under curve (AUC) value of receiver operating characteristic (ROC) curve was then used to assess the performance of the nomogram and independent predictors. RESULTS Among single region, intratumoral had the best predictive value. The diagnostic performance of the intratumoral improved after adding the BPE features. The AUC values of the radiomics signature were 0.822 and 0.82 in the training and validation sets. Multivariate logistic regression analysis revealed that age, ER, PR, Ki-67, and radiomics signature were independent predictors of pCR in constructing a nomogram. The AUC of the nomogram in the training and validation sets were 0.947 and 0.933. The DeLong test showed that the nomogram had statistically significant differences compared to other independent predictors in both the training and validation sets (P < 0.05). CONCLUSION BPE has value in predicting the efficacy of neoadjuvant chemotherapy, thereby revealing the potential impact of tumor growth environment on the efficacy of neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Guangying Zheng
- Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Jie Hou
- Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Zhenyu Shu
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Jiaxuan Peng
- Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Lu Han
- Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Zhongyu Yuan
- Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Xiaodong He
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Xiangyang Gong
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
4
|
Chaudhary LN, Jorns JM, Sun Y, Cheng YC, Kamaraju S, Burfeind J, Gonyo MB, Kong AL, Patten C, Yen T, Cortina CS, Carson E, Johnson N, Bergom C, Tsaih SW, Banerjee A, Wang Y, Chervoneva I, Weil E, Chitambar CR, Rui H. Frequent upregulation of HER2 protein in hormone-receptor-positive HER2-negative breast cancer after short-term neoadjuvant endocrine therapy. Breast Cancer Res Treat 2023; 201:387-396. [PMID: 37460683 PMCID: PMC10795510 DOI: 10.1007/s10549-023-07038-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Endocrine resistant metastatic disease develops in ~ 20-25% of hormone-receptor-positive (HR+) breast cancer (BC) patients despite endocrine therapy (ET) use. Upregulation of HER family receptor tyrosine kinases (RTKs) represent escape mechanisms in response to ET in some HR+ tumors. Short-term neoadjuvant ET (NET) offers the opportunity to identify early endocrine escape mechanisms initiated in individual tumors. METHODS This was a single arm, interventional phase II clinical trial evaluating 4 weeks (± 1 week) of NET in patients with early-stage HR+/HER2-negative (HER2-) BC. The primary objective was to assess NET-induced changes in HER1-4 proteins by immunohistochemistry (IHC) score. Protein upregulation was defined as an increase of ≥ 1 in IHC score following NET. RESULTS Thirty-seven patients with cT1-T3, cN0, HR+/HER2- BC were enrolled. In 35 patients with evaluable tumor HER protein after NET, HER2 was upregulated in 48.6% (17/35; p = 0.025), with HER2-positive status (IHC 3+ or FISH-amplified) detected in three patients at surgery, who were recommended adjuvant trastuzumab-based therapy. Downregulation of HER3 and/or HER4 protein was detected in 54.2% of tumors, whereas HER1 protein remained low and unchanged in all cases. While no significant volumetric reduction was detected radiographically after short-term NET, significant reduction in tumor proliferation rates were observed. No significant associations were identified between any clinicopathologic covariates and changes in HER1-4 protein expression on multivariable analysis. CONCLUSION Short-term NET frequently and preferentially upregulates HER2 over other HER family RTKs in early-stage HR+/HER2- BC and may be a promising strategy to identify tumors that utilize HER2 as an early endocrine escape pathway. CLINICAL TRIAL REGISTRY Trial registration number: NCT03219476.
Collapse
Affiliation(s)
- Lubna N Chaudhary
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA.
| | - Julie M Jorns
- Department of Pathology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Yunguang Sun
- Department of Pathology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Yee Chung Cheng
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Sailaja Kamaraju
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - John Burfeind
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Mary Beth Gonyo
- Department of Radiology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Amanda L Kong
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Caitlin Patten
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Tina Yen
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Chandler S Cortina
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Ebony Carson
- Clinical Trials Office, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Nedra Johnson
- Clinical Trials Office, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Shirng-Wern Tsaih
- Department of Obstetrics and Gynecology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Anjishnu Banerjee
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, USA
| | - Yu Wang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, USA
| | - Inna Chervoneva
- Division of Biostatistics, Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Elizabeth Weil
- Division of Pharmacy, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Christopher R Chitambar
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Hallgeir Rui
- Department of Pathology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| |
Collapse
|
5
|
Hurvitz SA, Bardia A, Quiroga V, Park YH, Blancas I, Alonso-Romero JL, Vasiliev A, Adamchuk H, Salgado M, Yardley DA, Berzoy O, Zamora-Auñón P, Chan D, Spera G, Xue C, Ferreira E, Badovinac Crnjevic T, Pérez-Moreno PD, López-Valverde V, Steinseifer J, Fernando TM, Moore HM, Fasching PA. Neoadjuvant palbociclib plus either giredestrant or anastrozole in oestrogen receptor-positive, HER2-negative, early breast cancer (coopERA Breast Cancer): an open-label, randomised, controlled, phase 2 study. Lancet Oncol 2023; 24:1029-1041. [PMID: 37657462 DOI: 10.1016/s1470-2045(23)00268-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/10/2023] [Accepted: 05/24/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND The development of more potent selective oestrogen receptor antagonists and degraders (SERDs) that can be orally administered could help to address the limitations of current endocrine therapies. We report the primary and final analyses of the coopERA Breast Cancer study, designed to test whether giredestrant, a highly potent, non-steroidal, oral SERD, would show a stronger anti-proliferative effect than anastrozole after 2 weeks for oestrogen receptor-positive, HER2-negative, untreated early breast cancer. METHODS In this open-label, randomised, controlled, phase 2 study, postmenopausal women were eligible if they were aged 18 years or older; had clinical T stage (cT)1c to cT4a-c (≥1·5 cm within cT1c) oestrogen receptor-positive, HER2-negative, untreated early breast cancer; an Eastern Cooperative Oncology Group performance status of 0-1; and baseline Ki67 score of at least 5%. The study was conducted at 59 hospital or clinic sites in 11 countries globally. Participants were randomly assigned (1:1) to giredestrant 30 mg oral daily or anastrozole 1 mg oral daily on days 1-14 (window-of-opportunity phase) via an interactive web-based system with permuted-block randomisation with block size of four. Randomisation was stratified by cT stage, baseline Ki67 score, and progesterone receptor status. A 16-week neoadjuvant phase comprised the same regimen plus palbociclib 125 mg oral daily on days 1-21 of a 28-day cycle, for four cycles. The primary endpoint was geometric mean relative Ki67 score change from baseline to week 2 in patients with complete central Ki67 scores at baseline and week 2 (window-of-opportunity phase). Safety was assessed in all patients who received at least one dose of study drug. The study is registered with ClinicalTrials.gov (NCT04436744) and is complete. FINDINGS Between Sept 4, 2020, and June 22, 2021, 221 patients were enrolled and randomly assigned to the giredestrant plus palbociclib group (n=112; median age 62·0 years [IQR 57·0-68·5]) or anastrozole plus palbociclib group (n=109; median age 62·0 [57·0-67·0] years). 15 (7%) of 221 patients were Asian, three (1%) were Black or African American, 194 (88%) were White, and nine (4%) were unknown races. At data cutoff for the primary analysis (July 19, 2021), the geometric mean relative reduction of Ki67 from baseline to week 2 was -75% (95% CI -80 to -70) with giredestrant and -67% (-73 to -59) with anastrozole (p=0·043), meeting the primary endpoint. At the final analysis (data cutoff Nov 24, 2021), the most common grade 3-4 adverse events were neutropenia (29 [26%] of 112 in the giredestrant plus palbociclib group vs 29 [27%] of 109 in the anastrozole plus palbociclib group) and decreased neutrophil count (17 [15%] vs 16 [15%]). Serious adverse events occurred in five (4%) patients in the giredestrant plus palbociclib group and in two (2%) patients in the anastrozole plus palbociclib group. There were no treatment-related deaths. One patient died due to an adverse event in the giredestrant plus palbociclib group (myocardial infarction). INTERPRETATION Giredestrant offers encouraging anti-proliferative and anti-tumour activity and was well tolerated, both as a single agent and in combination with palbociclib. Results justify further investigation in ongoing trials. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Sara A Hurvitz
- Breast Cancer Clinical Trials Program, Division of Hematology-Oncology, David Geffen School of Medicine, Clinical Research Unit, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA; Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Aditya Bardia
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vanesa Quiroga
- GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain; Catalan Institute of Oncology Badalona, Barcelona, Spain
| | | | - Isabel Blancas
- GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain; Hospital Universitario San Cecilio, Instituto de Investigación Biosanitaria de Granada, Granada, Spain; Medicine Department, Granada University, Granada, Spain
| | - José Luis Alonso-Romero
- GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain; Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain
| | - Aleksandr Vasiliev
- NSHI Road Clinical Hospital of JSC Russian Railways, Saint Petersburg, Russia
| | - Hryhoriy Adamchuk
- Communal Enterprise Kryvyi Rih Oncology Dispensary, Kryvyi Rih, Ukraine
| | | | - Denise A Yardley
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN, USA
| | - Oleksandr Berzoy
- Communal Non-profit Enterprise Odesa Regional Clinical Hospital of Odesa Regional Council, Odesa, Ukraine
| | - Pilar Zamora-Auñón
- GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain; Hospital Universitario La Paz, Madrid, Spain
| | - David Chan
- Torrance Memorial Hunt Cancer Center, Torrance, CA, USA
| | - Gonzalo Spera
- Translational Research in Oncology (TRIO), Montevideo, Uruguay
| | - Cloris Xue
- F Hoffmann-La Roche, Toronto, ON, Canada
| | | | | | | | | | | | | | | | - Peter A Fasching
- University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
6
|
Gnant M, Turner NC, Hernando C. Managing a Long and Winding Road: Estrogen Receptor-Positive Breast Cancer. Am Soc Clin Oncol Educ Book 2023; 43:e390922. [PMID: 37319380 DOI: 10.1200/edbk_390922] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
We review key topics in the management of estrogen receptor (ER)-positive human epidermal growth factor receptor 2-negative breast cancer. The single biggest challenge in management of this disease is late relapse, and we review new methods for identifying which patients are at risk of late relapse and potential therapeutic approaches in clinical trials. CDK4/6 inhibitors have become a standard treatment option for high-risk patients in both the adjuvant setting and the first-line metastatic setting, and we review data on optimal treatment after progression on CDK4/6 inhibitors. Targeting the estrogen receptor remains the single most effective way of targeting the cancer, and we review the developments in new oral selective ER degraders that are becoming a standard of care in cancers with ESR1 mutations and potential future directions.
Collapse
Affiliation(s)
- Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria
| | - Nicholas C Turner
- The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Cristina Hernando
- Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, Valencia, Spain
| |
Collapse
|
7
|
Chaudhary LN, Jorns J, Sun Y, Cheng YC, Kamaraju S, Burfeind J, Gonyo M, Kong A, Patten C, Yen T, Cortina C, Carson E, Johnson N, Bergom C, Tsaih SW, Banerjee A, Wang Y, Chervoneva I, Weil E, Chitambar CR, Rui H. Frequent Upregulation Of HER2 Protein In Hormone Receptor-Positive HER2-Negative Breast Cancer After Short-Term Neoadjuvant Endocrine Therapy. RESEARCH SQUARE 2023:rs.3.rs-2777910. [PMID: 37066270 PMCID: PMC10104267 DOI: 10.21203/rs.3.rs-2777910/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Background. Endocrine resistant metastatic disease develops in ~20-25% of hormone-receptor positive (HR+) breast cancer (BC) patients despite endocrine therapy (ET) use. Upregulation of HER family receptor tyrosine kinases (RTKs) represent escape mechanisms in response to ET in some HR+ tumors. Short-term neoadjuvant ET (NET) offers the opportunity to identify early endocrine escape mechanisms initiated in individual tumors. Methods. This was a single arm, interventional phase II clinical trial evaluating 4 weeks (+/-1 week) of NET in patients with early-stage HR+/HER2-negative (HER2-) BC. The primary objective was to assess NET-induced changes in HER1-4 proteins by immunohistochemistry (IHC) score. Protein upregulation was defined as an increase of ≥1 in IHC score following NET. Results. Thirty-seven patients with cT1-T3, cN0, HR+/HER2- BC were enrolled. In 35 patients with evaluable tumor HER protein after NET, HER2 was upregulated in 48.6% (17/35; p=0.025), with HER2-positive status (IHC 3+ or FISH-amplified) detected in three patients at surgery, who were recommended adjuvant trastuzumab-based therapy. Downregulation of HER3 and/or HER4 protein was detected in 54.2% of tumors, whereas HER1 protein remained low and unchanged in all cases. While no significant volumetric reduction was detected radiographically after short-term NET, significant reduction in tumor proliferation rates were observed. No significant associations were identified between any clinicopathologic covariates and changes in HER1-4 protein expression on multivariable analysis. Conclusion . Short-term NET frequently and preferentially upregulates HER2 over other HER-family RTKs in early-stage HR+/HER2- BC and may be a promising strategy to identify tumors that utilize HER2 as an early endocrine escape pathway. Trial registration number: NCT03219476 Date of registration for prospectively registered trials: July 17, 2017.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tina Yen
- MCW: Medical College of Wisconsin
| | | | | | | | | | | | | | - Yu Wang
- MCW: Medical College of Wisconsin
| | | | | | | | | |
Collapse
|
8
|
Louis DM, Nair LM, Vallonthaiel AG, Narmadha MP, Vijaykumar DK. Ki 67: a Promising Prognostic Marker in Early Breast Cancer-a Review Article. Indian J Surg Oncol 2023; 14:122-127. [PMID: 36891414 PMCID: PMC9986372 DOI: 10.1007/s13193-022-01631-6] [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: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022] Open
Abstract
Ki67 index is considered to be a reliable indicator of the proliferative activity of breast cancer. Additionally, the Ki67 proliferative marker may play a role in assessing response to systemic therapeutic strategies and can act as a prognostic biomarker. But its limited reproducibility which stems from a lack of standardization of procedures, inter-observer variability, and preanalytical and analytical variabilities all have hampered the use of the Ki67 index in clinical practice. Currently, clinical trials have been evaluating Ki67 as a predictive marker for needing adjuvant chemotherapy in luminal early breast cancer patients receiving neoadjuvant endocrine therapy. But the inconsistencies existing in the estimation of the Ki67 index limit the utility of Ki67 in standard clinical practice. The purpose of this review is to evaluate the benefits and drawbacks of utilizing Ki-67 in early-stage breast cancer to prognosticate the disease and predict the risk of recurrence.
Collapse
Affiliation(s)
- Dhanya Mary Louis
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Institute of Medical Science and Research Centre, AIMS-Ponekkara, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041 India
| | - Lakshmi Malavika Nair
- Breast Diseases Division, Amrita School of Medicine, Amrita Institute of Medical Science and Research Centre, AIMS-Ponekkara, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041 India
| | - Archana George Vallonthaiel
- Department of Pathology, Amrita School of Medicine, Amrita Institute of Medical Science and Research Centre, AIMS-Ponekkara, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041 India
| | - M. P. Narmadha
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Institute of Medical Science and Research Centre, AIMS-Ponekkara, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041 India
| | - D. K. Vijaykumar
- Breast Diseases Division, Amrita School of Medicine, Amrita Institute of Medical Science and Research Centre, AIMS-Ponekkara, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041 India
| |
Collapse
|
9
|
Martínez-Pérez C, Turnbull AK, Kay C, Dixon JM. Neoadjuvant endocrine therapy in postmenopausal women with HR+/HER2- breast cancer. Expert Rev Anticancer Ther 2023; 23:67-86. [PMID: 36633402 DOI: 10.1080/14737140.2023.2162043] [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: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
INTRODUCTION While endocrine therapy is the standard-of-care adjuvant treatment for hormone receptor-positive (HR+) breast cancers, there is also extensive evidence for the role of pre-operative (or neoadjuvant) endocrine therapy (NET) in HR+ postmenopausal women. AREAS COVERED We conducted a thorough review of the published literature, to summarize the evidence to date, including studies of how NET compares to neoadjuvant chemotherapy, which NET agents are preferable, and the optimal duration of NET. We describe the importance of on-treatment assessment of response, the different predictors available (including Ki67, PEPI score, and molecular signatures) and the research opportunities the pre-operative setting offers. We also summarize recent combination trials and discuss how the COVID-19 pandemic led to increases in NET use for safe management of cases with deferred surgery and adjuvant treatments. EXPERT OPINION NET represents a safe and effective tool for the management of postmenopausal women with HR+/HER2- breast cancer, enabling disease downstaging and a wider range of surgical options. Aromatase inhibitors are the preferred NET, with evidence suggesting that longer regimens might yield optimal results. However, NET remains currently underutilised in many territories and institutions. Further validation of predictors for treatment response and benefit is needed to help standardise and fully exploit the potential of NET in the clinic.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Arran K Turnbull
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Charlene Kay
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - J Michael Dixon
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, Scotland
| |
Collapse
|
10
|
Calaf GM, Crispin LA, Muñoz JP, Aguayo F, Bleak TC. Muscarinic Receptors Associated with Cancer. Cancers (Basel) 2022; 14:cancers14092322. [PMID: 35565451 PMCID: PMC9100020 DOI: 10.3390/cancers14092322] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Recently, cancer research has described the presence of the cholinergic machinery, specifically muscarinic receptors, in a wide variety of cancers due to their activation and signaling pathways associated with tumor progression and metastasis, providing a wide overview of their contribution to different cancer formation and development for new antitumor targets. This review focused on determining the molecular signatures associated with muscarinic receptors in breast and other cancers and the need for pharmacological, molecular, biochemical, technological, and clinical approaches to improve new therapeutic targets. Abstract Cancer has been considered the pathology of the century and factors such as the environment may play an important etiological role. The ability of muscarinic agonists to stimulate growth and muscarinic receptor antagonists to inhibit tumor growth has been demonstrated for breast, melanoma, lung, gastric, colon, pancreatic, ovarian, prostate, and brain cancer. This work aimed to study the correlation between epidermal growth factor receptors and cholinergic muscarinic receptors, the survival differences adjusted by the stage clinical factor, and the association between gene expression and immune infiltration level in breast, lung, stomach, colon, liver, prostate, and glioblastoma human cancers. Thus, targeting cholinergic muscarinic receptors appears to be an attractive therapeutic alternative due to the complex signaling pathways involved.
Collapse
Affiliation(s)
- Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
- Correspondence:
| | - Leodan A. Crispin
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| | - Juan P. Muñoz
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| | - Francisco Aguayo
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Tammy C. Bleak
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| |
Collapse
|
11
|
Lopez-Tarruella S, Echavarria I, Jerez Y, Herrero B, Gamez S, Martin M. How we treat HR-positive, HER2-negative early breast cancer. Future Oncol 2022; 18:1003-1022. [PMID: 35094535 DOI: 10.2217/fon-2021-0668] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The present goal of therapy for early hormone receptor-positive (HR+)/human EGF receptor 2-negative (HER2-) BC is to optimize disease-free survival (DFS) and overall survival (OS) rates with the currently available therapies while avoiding any relevant long-term sequalae. Local therapies have evolved toward less aggressive techniques (i.e. breast-preserving surgery, sentinel lymph node biopsy and intraoperative radiotherapy), which significantly reduce the long-term sequalae observed with more radical treatments. Endocrine therapy (ET) is still the cornerstone of adjuvant treatment because it significantly reduces BC relapse and mortality. Adjuvant chemotherapy is today recommended only for a particular subset of patients with a high risk of recurrence with ET alone, identified through genomic assays, age and/or disease stage. Bisphosphonates reduce the risk of bone metastasis and produce a slight although statistically significant improvement in survival in postmenopausal women. The CDK 4/6 inhibitor abemaciclib has been recently approved by the US FDA for patients at high risk of relapse.
Collapse
Affiliation(s)
- Sara Lopez-Tarruella
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, CiberOnc, GEICAM, Madrid, 28007, Spain
| | - Isabel Echavarria
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, 28007, Spain
| | - Yolanda Jerez
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, CiberOnc, GEICAM, Madrid, 28007, Spain
| | - Blanca Herrero
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, 28007, Spain
| | - Salvador Gamez
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, 28007, Spain
| | - Miguel Martin
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, CiberOnc, GEICAM, Madrid, 28007, Spain
| |
Collapse
|
12
|
Clinical Translation: Targeting the Estrogen Receptor. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:297-309. [DOI: 10.1007/978-3-031-11836-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Antibiotics modulate neoadjuvant therapy efficiency in patients with breast cancer: a pilot analysis. Sci Rep 2021; 11:14024. [PMID: 34234229 PMCID: PMC8263554 DOI: 10.1038/s41598-021-93428-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/22/2021] [Indexed: 11/08/2022] Open
Abstract
Mounting evidence suggests that microbiota dysbiosis caused by antibiotic administration is a risk factor for cancer, but few research reports focus on the relationships between antibiotics and chemotherapy efficiency. We evaluated the influence of antibiotic administration on neoadjuvant therapy efficacy in patients with breast cancer (BC) in the present study. BC patients were stratified into two groups: antibiotic-treated and control based on antibiotic administration within 30 days after neoadjuvant therapy initiation. Disease-free survival (DFS) and overall survival (OS) were assessed using the Kaplan-Meier method, and the Cox proportional hazards model was used for multivariate analyses. The pathologic complete response rate of the control group was significantly higher than that of the antibiotic-treated group (29.09% vs. 10.20%, p = 0.017). Further univariate analysis with Kaplan-Meier calculations demonstrated that antibiotic administration was strongly linked with both reduced DFS (p = 0.04) at significant statistical levels and OS (p = 0.088) at borderline statistical levels. Antibiotic administration was identified as a significant independent prognostic factor for DFS [hazard ratio (HR) 3.026, 95%, confidence interval (CI) 1.314-6.969, p = 0.009] and OS (HR 2.836, 95% CI 1.016-7.858, p = 0.047) by Cox proportional hazards model analysis. Antibiotics that initiated reduced efficiency of chemotherapy were more noticeable in the HER2-positive subgroup for both DFS (HR 5.51, 95% CI 1.77-17.2, p = 0.003) and OS (HR 7.0395% CI 1.94-25.53, p = 0.003), as well as in the T3-4 subgroup for both DFS (HR 20.36, 95% CI 2.41-172.07, p = 0.006) and OS (HR 13.45, 95% CI 1.39-130.08, p = 0.025) by stratified analysis. Antibiotic administration might be associated with reduced efficacy of neoadjuvant therapy and poor prognosis in BC patients. As a preliminary study, our research made preparations for further understanding and large-scale analyses of the impact of antibiotics on the efficacy of neoadjuvant therapy.
Collapse
|
14
|
The Present and Future of Neoadjuvant Endocrine Therapy for Breast Cancer Treatment. Cancers (Basel) 2021; 13:cancers13112538. [PMID: 34064183 PMCID: PMC8196711 DOI: 10.3390/cancers13112538] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The treatment of breast cancer has evolved considerably over the last two decades, leading toward individualized disease management. Hormone-sensitive breast cancers constitute the vast majority of cases and endocrine therapy is the mainstay of their treatment. On the other hand, neoadjuvant or pre-surgical treatments provide a number of advantages for tumor management. In this review we will discuss the existing evidence on neoadjuvant endocrine therapy, as well as its possible future indications. Abstract Endocrine therapy (ET) has established itself as an efficacious treatment for estrogen receptor-positive (ER+) breast cancers, with a reduction in recurrence rates and increased survival rates. The pre-surgical approach with chemotherapy (NCT) has become a common form of management for large, locally advanced, or high-risk tumors. However, a good response to NCT is not usually expected in ER+ tumors. Good results with primary ET, mainly in elderly women, have encouraged studies in other stages of life, and nowadays neoadjuvant endocrine treatment (NET) has become a useful approach to many ER+ breast cancers. The aim of this review is to provide an update on the current state of art regarding the present and the future role of NET.
Collapse
|
15
|
Ashirbekov Y, Abaildayev A, Omarbayeva N, Botbayev D, Belkozhayev A, Askandirova A, Neupokoyeva A, Utegenova G, Sharipov K, Aitkhozhina N. Combination of circulating miR-145-5p/miR-191-5p as biomarker for breast cancer detection. PeerJ 2020; 8:e10494. [PMID: 33362968 PMCID: PMC7749656 DOI: 10.7717/peerj.10494] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background Breast cancer (BC) is the most common cancer among women worldwide. At present, there is a need to search for new, accurate, reliable, minimally invasive and cheap biomarkers in addition to existing methods for the diagnosis and prognosis of BC. The main goal of this study was to test the diagnostic value of six circulating miRNAs in Kazakh women. Materials and methods TaqMan-based miRNA profiling was conducted using plasma specimens from 35 BC women patients and 33 healthy women samples (control group). Results The level of all seven miRNAs (including endogenous control) normalized by synthetic cel-miR-39 were significantly elevated in the group of BC patients. Normalization using miR-222-3p as endogenous control reduced differences in level of miRNAs between groups; as a result, only three miRNAs were significantly upregulated in the group of BC patients—miR-145-5p (P = 6.5e−12), miR-191-5p (P = 3.7e−10) and miR-21-5p (P = 0.0034). Moreover, ROC analysis showed that the use of miR-145-5p and miR-191-5p, both individually (AUC = 0.931 and 0.904, respectively) or in combination (AUC = 0.984), allows to accurately differentiate BC patients from healthy individuals. Conclusions Two plasma miRNAs—miR-145-5p and miR-191-5p—are potential biomarkers for diagnosis of BC in the Kazakh population. The findings need to be further substantiated using a more representative sample.
Collapse
Affiliation(s)
- Yeldar Ashirbekov
- M. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Arman Abaildayev
- M. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Nazgul Omarbayeva
- Kazakh Research Institute of Oncology and Radiology, Almaty, Kazakhstan
| | - Dauren Botbayev
- M. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Ayaz Belkozhayev
- M. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Anel Askandirova
- Kazakh Research Institute of Oncology and Radiology, Almaty, Kazakhstan
| | - Alena Neupokoyeva
- Almaty Branch of National Center for Biotechnology, Almaty, Kazakhstan
| | | | - Kamalidin Sharipov
- M. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Nagima Aitkhozhina
- M. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| |
Collapse
|
16
|
Jones EF, Hathi DK, Freimanis R, Mukhtar RA, Chien AJ, Esserman LJ, van’t Veer LJ, Joe BN, Hylton NM. Current Landscape of Breast Cancer Imaging and Potential Quantitative Imaging Markers of Response in ER-Positive Breast Cancers Treated with Neoadjuvant Therapy. Cancers (Basel) 2020; 12:E1511. [PMID: 32527022 PMCID: PMC7352259 DOI: 10.3390/cancers12061511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Abstract
In recent years, neoadjuvant treatment trials have shown that breast cancer subtypes identified on the basis of genomic and/or molecular signatures exhibit different response rates and recurrence outcomes, with the implication that subtype-specific treatment approaches are needed. Estrogen receptor-positive (ER+) breast cancers present a unique set of challenges for determining optimal neoadjuvant treatment approaches. There is increased recognition that not all ER+ breast cancers benefit from chemotherapy, and that there may be a subset of ER+ breast cancers that can be treated effectively using endocrine therapies alone. With this uncertainty, there is a need to improve the assessment and to optimize the treatment of ER+ breast cancers. While pathology-based markers offer a snapshot of tumor response to neoadjuvant therapy, non-invasive imaging of the ER disease in response to treatment would provide broader insights into tumor heterogeneity, ER biology, and the timing of surrogate endpoint measurements. In this review, we provide an overview of the current landscape of breast imaging in neoadjuvant studies and highlight the technological advances in each imaging modality. We then further examine some potential imaging markers for neoadjuvant treatment response in ER+ breast cancers.
Collapse
Affiliation(s)
- Ella F. Jones
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Deep K. Hathi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Rita Freimanis
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Rita A. Mukhtar
- Department of Surgery, University of California, San Francisco, CA 94115, USA;
| | - A. Jo Chien
- School of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (A.J.C.); (L.J.v.V.)
| | - Laura J. Esserman
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA;
| | - Laura J. van’t Veer
- School of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (A.J.C.); (L.J.v.V.)
| | - Bonnie N. Joe
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Nola M. Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| |
Collapse
|
17
|
Geread RS, Morreale P, Dony RD, Brouwer E, Wood GA, Androutsos D, Khademi A. IHC Color Histograms for Unsupervised Ki67 Proliferation Index Calculation. Front Bioeng Biotechnol 2019; 7:226. [PMID: 31632956 PMCID: PMC6779686 DOI: 10.3389/fbioe.2019.00226] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/03/2019] [Indexed: 12/23/2022] Open
Abstract
Automated image analysis tools for Ki67 breast cancer digital pathology images would have significant value if integrated into diagnostic pathology workflows. Such tools would reduce the workload of pathologists, while improving efficiency, and accuracy. Developing tools that are robust and reliable to multicentre data is challenging, however, differences in staining protocols, digitization equipment, staining compounds, and slide preparation can create variabilities in image quality and color across digital pathology datasets. In this work, a novel unsupervised color separation framework based on the IHC color histogram (IHCCH) is proposed for the robust analysis of Ki67 and hematoxylin stained images in multicentre datasets. An "overstaining" threshold is implemented to adjust for background overstaining, and an automated nuclei radius estimator is designed to improve nuclei detection. Proliferation index and F1 scores were compared between the proposed method and manually labeled ground truth data for 30 TMA cores that have ground truths for Ki67+ and Ki67- nuclei. The method accurately quantified the PI over the dataset, with an average proliferation index difference of 3.25%. To ensure the method generalizes to new, diverse datasets, 50 Ki67 TMAs from the Protein Atlas were used to test the validated approach. As the ground truth for this dataset is PI ranges, the automated result was compared to the PI range. The proposed method correctly classified 74 out of 80 TMA images, resulting in a 92.5% accuracy. In addition to these validations experiments, performance was compared to two color-deconvolution based methods, and to six machine learning classifiers. In all cases, the proposed work maintained more consistent (reproducible) results, and higher PI quantification accuracy.
Collapse
Affiliation(s)
- Rokshana S Geread
- Image Analysis in Medicine Lab, Ryerson University, Toronto, ON, Canada
| | - Peter Morreale
- School of Engineering, University of Guelph, Guelph, ON, Canada
| | - Robert D Dony
- School of Engineering, University of Guelph, Guelph, ON, Canada
| | - Emily Brouwer
- Ontario Veterinarian College, University of Guelph, Guelph, ON, Canada
| | - Geoffrey A Wood
- Ontario Veterinarian College, University of Guelph, Guelph, ON, Canada
| | | | - April Khademi
- Image Analysis in Medicine Lab, Ryerson University, Toronto, ON, Canada
| |
Collapse
|
18
|
Adamo B, Bellet M, Paré L, Pascual T, Vidal M, Pérez Fidalgo JA, Blanch S, Martinez N, Murillo L, Gómez-Pardo P, López-González A, Amillano K, Canes J, Galván P, González-Farré B, González X, Villagrasa P, Ciruelos E, Prat A. Oral metronomic vinorelbine combined with endocrine therapy in hormone receptor-positive HER2-negative breast cancer: SOLTI-1501 VENTANA window of opportunity trial. Breast Cancer Res 2019; 21:108. [PMID: 31533777 PMCID: PMC6751874 DOI: 10.1186/s13058-019-1195-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 09/02/2019] [Indexed: 12/17/2022] Open
Abstract
Background The biological effect of oral metronomic vinorelbine (mVNB) alone or in combination with endocrine therapy in patients with hormone receptor-positive (HR+)/HER2-negative breast cancer has been scarcely addressed. Methods Postmenopausal women with untreated stage I–III HR+/HER2-negative breast cancer were randomized (1:1:1) to receive 3 weeks of letrozole (LTZ) 2.5 mg/day, oral mVNB 50 mg 3 days/week, or the combination. The primary objective was to evaluate, within PAM50 Luminal A/B disease, if the anti-proliferative effect of LTZ+mVNB was superior to monotherapy. An anti-proliferative effect was defined as the mean relative decrease of the PAM50 11-gene proliferation score in combination arm vs. both monotherapy arms. Secondary objectives included the evaluation of a comprehensive panel of breast cancer-related genes and safety. An unplanned analysis of stromal tumor-infiltrating lymphocytes (sTILs) was also performed. PAM50 analyses were performed using the nCounter®-based Breast Cancer 360™ gene panel, which includes 752 genes and 32 signatures. Results Sixty-one patients were randomized, and 54 paired samples (89%) were analyzed. The main patient characteristics were mean age of 67, mean tumor size of 1.7 cm, mean Ki67 of 14.3%, stage I (55.7%), and grades 1–2 (90%). Most baseline samples were PAM50 Luminal A (74.1%) or B (22.2%). The anti-proliferative effect of 3 weeks of LTZ+mVNB (− 73.2%) was superior to both monotherapy arms combined (− 49.9%; p = 0.001) and mVNB (− 19.1%; p < 0.001). The anti-proliferative effect of LTZ+mVNB (− 73.2%) was numerically higher compared to LTZ (− 65.7%) but did not reach statistical significance (p = 0.328). LTZ+mVNB induced high expression of immune-related genes and gene signatures, including CD8 T cell signature and PDL1 gene and low expression of ER-regulated genes (e.g., progesterone receptor) and cell cycle-related and DNA repair genes. In tumors with ≤ 10% sTILs at baseline, a statistically significant increase in sTILs was observed following LTZ (paired analysis p = 0.049) and LTZ+mVNB (p = 0.012). Grade 3 adverse events occurred in 3.4% of the cases. Conclusions Short-term mVNB is well-tolerated and presents anti-proliferative activity alone and in combination with LTZ. The high expression of immune-related biological processes and sTILs observed with the combination opens the possibility of studying this combination with immunotherapy. Further investigation comparing these biological results with other metronomic schedules or drug combinations is warranted. Trial registration NCT02802748, registered 16 June 2016. Supplementary information Supplementary information accompanies this paper at 10.1186/s13058-019-1195-z.
Collapse
Affiliation(s)
- Barbara Adamo
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Villarroel 170, 08035, Barcelona, Spain
| | - Meritxell Bellet
- Vall d'Hebrón University Hospital/Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Laia Paré
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Villarroel 170, 08035, Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Tomás Pascual
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Villarroel 170, 08035, Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Maria Vidal
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Villarroel 170, 08035, Barcelona, Spain
| | | | - Salvador Blanch
- Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | | | - Laura Murillo
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Patricia Gómez-Pardo
- Vall d'Hebrón University Hospital/Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | - Jordi Canes
- SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Patricia Galván
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | | | | | - Eva Ciruelos
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Aleix Prat
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain. .,Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Villarroel 170, 08035, Barcelona, Spain. .,SOLTI Breast Cancer Research Group, Barcelona, Spain.
| |
Collapse
|
19
|
Marra A, Curigliano G. Are all cyclin-dependent kinases 4/6 inhibitors created equal? NPJ Breast Cancer 2019; 5:27. [PMID: 31482107 PMCID: PMC6715721 DOI: 10.1038/s41523-019-0121-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
The harnessing in clinical practice of cyclin-dependent kinases 4/6 inhibitors, namely palbociclib, ribociclib, and abemaciclib, has substantially changed the therapeutic approach for hormone receptor-positive metastatic breast cancer (BC). Phase II-III clinical trials evaluating the addition of these agents to standard endocrine therapy reported consistent improvements in response rates and progression-free survival as well as manageable toxicity profiles and excellent impact on patients' quality of life. Hence, pivotal trials provided comparable results among different cyclin-dependent kinases 4/6 inhibitors, there is an increasing interest in finding substantial differences in order to implement their use in clinical practice. The aim of this paper is to summarize the current evidences raised from preclinical and clinical studies on cyclin-dependent kinases 4/6 inhibitors in BC, focusing on differences in terms of pharmacological properties, toxicity profile, and patients' quality of life.
Collapse
Affiliation(s)
- Antonio Marra
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| |
Collapse
|
20
|
Saura C, Hlauschek D, Oliveira M, Zardavas D, Jallitsch-Halper A, de la Peña L, Nuciforo P, Ballestrero A, Dubsky P, Lombard JM, Vuylsteke P, Castaneda CA, Colleoni M, Santos Borges G, Ciruelos E, Fornier M, Boer K, Bardia A, Wilson TR, Stout TJ, Hsu JY, Shi Y, Piccart M, Gnant M, Baselga J, de Azambuja E. Neoadjuvant letrozole plus taselisib versus letrozole plus placebo in postmenopausal women with oestrogen receptor-positive, HER2-negative, early-stage breast cancer (LORELEI): a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 2019; 20:1226-1238. [PMID: 31402321 DOI: 10.1016/s1470-2045(19)30334-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Endocrine therapy-based neoadjuvant treatment for luminal breast cancer allows efficient testing of new combinations before surgery. The activation of the phosphatidylinositol-3-kinase (PI3K) pathway is a known mechanism of resistance to endocrine therapy. Taselisib is an oral, selective PI3K inhibitor with enhanced activity against PIK3CA-mutant cancer cells. The LORELEI trial tested whether taselisib in combination with letrozole would result in an increased proportion of objective responses and pathological complete responses. METHODS In this multicentre, randomised, double-blind, parallel-cohort, placebo-controlled phase 2, study, we enrolled postmenopausal women (aged ≥18 years) with histologically confirmed, oestrogen receptor (ER)-positive, HER2-negative, stage I-III, operable breast cancer, from 85 hospitals in 22 countries worldwide. To be eligible, patients had have an Eastern Cooperative Oncology Group (ECOG) performance status 0-1, adequate organ function, and had to have evaluable tumour tissue for PIK3CA genotyping. Patients were randomly assigned (1:1) by means of a permuted block algorithm (block size of four) via an interactive voice or web-based response system, to receive letrozole (2·5 mg/day orally, continuously) with either 4 mg of oral taselisib or placebo (on a 5 days-on, 2 days-off schedule) for 16 weeks, followed by surgery. Randomisation was stratified by tumour size and nodal status. Site staff, patients, and the sponsor were masked to treatment assignment. Coprimary endpoints were the proportion of patients who achieved an objective response by centrally assessed breast MRI and a locally assessed pathological complete response in the breast and axilla (ypT0/Tis, ypN0) at surgery in all randomly assigned patients and in patients with PIK3CA-mutant tumours. Analyses were done in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, number NCT02273973, and is closed to accrual. FINDINGS Between Nov 12, 2014, and Aug 12, 2016, 334 participants were enrolled and randomly assigned to receive letrozole and placebo (n=168) or letrozole and taselisib (n=166). Median follow-up was 4·9 months (IQR 4·7-5·1). The study met one of its primary endpoints: the addition of taselisib to letrozole was associated with a higher proportion of patients achieving an objective response in all randomly assigned patients (66 [39%] of 168 patients in the placebo group vs 83 [50%] of 166 in the taselisib group; odds ratio [OR] 1·55, 95% CI 1·00-2·38; p=0·049) and in the PIK3CA-mutant subset (30 [38%] of 79 vs 41 [56%] of 73; OR 2·03, 95% CI 1·06-3·88; p=0·033). No significant differences were observed in pathological complete response between the two groups, either in the overall population (three [2%] of 166 in the taselisib group vs one [1%] of 168 in the placebo group; OR 3·07 [95% CI 0·32-29·85], p=0·37) or in the PIK3CA-mutant cohort (one patient [1%) vs none [0%]; OR not estimable, p=0·48). The most common grade 3-4 adverse events in the taselisib group were gastrointestinal (13 [8%] of 167 patients), infections (eight [5%]), and skin-subcutaneous tissue disorders (eight [5%]). In the placebo group, four (2%) of 167 patients had grade 3 or worse vascular disorders, two (1%) had gastrointestinal disorders, and two (1%) patients had grade 3 or worse infections and infestations. There was no grade 4 hyperglycaemia and grade 3 cases were asymptomatic. Serious adverse events were more common in the taselisib group (eight [5%] patients with infections and seven [4%] with gastrointestinal effects) than in the placebo group (one [1%] patient each with grade 3 postoperative wound and haematoma infection, grade 4 hypertensive encephalopathy, grade 3 acute cardiac failure, and grade 3 breast pain). One death occurred in the taselisib group, which was not considered to be treatment-related. INTERPRETATION The increase in the proportion of patients who achieved an objective response from the addition of taselisib to endocrine therapy in a neoadjuvant setting is consistent with the clinical benefit observed in hormone receptor-positive, HER2-negative, metastatic breast cancer. FUNDING Genentech and F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Cristina Saura
- Vall d'Hebrón University Hospital, Vall d'Hebrón Institute of Oncology, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain.
| | | | - Mafalda Oliveira
- Vall d'Hebrón University Hospital, Vall d'Hebrón Institute of Oncology, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain
| | | | | | | | - Paolo Nuciforo
- Vall d'Hebrón University Hospital, Vall d'Hebrón Institute of Oncology, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Alberto Ballestrero
- University of Genoa - Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Policlinico San Martino, Genova; Gruppo Oncologico Italiano di Ricerca Clinica, Genova, Italy
| | - Peter Dubsky
- Medical University of Vienna, Vienna, Austria; Hirslanden Klinik St Anna, Breast Centre, Lucerne, Switzerland
| | - Janine M Lombard
- Breast Cancer Trials Australia New Zealand, University of Newcastle, Newcastle, NSW, Australia
| | - Peter Vuylsteke
- European Organisation for Research and Treatment of Cancer, Breast Cancer Group, CHU Namur, Université Catholique de Louvain, Ottignies-Nouvain-la-Neuve, Belgium
| | | | - Marco Colleoni
- Division of Medical Senology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico and International Breast Cancer Study Group, Milan, Italy
| | | | - Eva Ciruelos
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Monica Fornier
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | | | - Aditya Bardia
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Yi Shi
- Genentech Inc, South San Francisco, CA, USA
| | - Martine Piccart
- Breast International Group, Brussels, Belgium; Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Michael Gnant
- Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria; Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
21
|
Sato N, Masuda N, Morimoto T, Ueno T, Kanbayashi C, Kaneko K, Yasojima H, Saji S, Sasano H, Morita S, Ohno S, Toi M. Neoadjuvant exemestane or exemestane plus docetaxel and cyclophosphamide tailored by clinicopathological response to 12 weeks' exemestane exposure in patients with estrogen receptor-positive breast cancer: A multicenter, open-label, phase II study. Cancer Med 2019; 8:5468-5481. [PMID: 31361400 PMCID: PMC6745863 DOI: 10.1002/cam4.2423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/21/2022] Open
Abstract
Our aim was to investigate the efficacy and safety of initial neoadjuvant endocrine therapy with exemestane alone followed by tailored treatment, either continued exemestane monotherapy or exemestane plus docetaxel–cyclophosphamide (TC) combination therapy, in postmenopausal patients with primary invasive estrogen receptor–positive, human epidermal growth factor receptor 2–negative, stage I‐IIIA breast cancer and Ki67 labeling index ≤30%. In this open‐label phase II study, patients initially received exemestane 25 mg/d for 12 weeks. Responders were defined as patients who achieved complete response (CR), partial response (PR) with Ki67 labeling index ≤5% after treatment, or stable disease with Ki67 labeling index ≤5% both before and after treatment. For the subsequent 12 weeks, exemestane monotherapy was continued for responders (group A), whereas nonresponders received exemestane plus four cycles of TC (docetaxel 75 mg/m2 and cyclophosphamide 600 mg/m2 every 3 weeks) (group B). Clinical response rate (ie the proportion of patients with CR or PR) at 24 weeks was the primary endpoint. Of 64 patients provisionally enrolled between December 2010 and May 2016, 58 (median age 60 years) started the study treatment. Five patients discontinued treatment in the initial exemestane monotherapy period, and 39 completed the study treatment. Clinical response rates at 8‐12 and 24 weeks were 71% (10/14, 95% confidence interval [CI] 41.9%‐91.6%) and 57% (8/14, 95% CI 28.9%‐82.3%), respectively, in group A, and 16% (4/25, 95% CI 4.5%‐36.1%) and 56% (14/25, 95% CI 34.9%‐75.6%), respectively, in group B. Grade ≥3 adverse events were reported in 8% (1/15) and 53% (20/38) in group A and group B, respectively. The tailored treatment maintained the favorable clinical response to exemestane alone in responders and improved clinical response in nonresponders. Trial number UMIN000004752 (UMIN Clinical Trials Registry).
Collapse
Affiliation(s)
- Nobuaki Sato
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Norikazu Masuda
- Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Takashi Morimoto
- Department of Breast Surgery, Yao Municipal Hospital, Osaka, Japan
| | - Takayuki Ueno
- Breast Surgical Oncology, Breast Oncology Center, Cancer Institute Hospital, Tokyo, Japan
| | - Chizuko Kanbayashi
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Koji Kaneko
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hiroyuki Yasojima
- Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | | | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinji Ohno
- Breast Oncology Center, Cancer Institute Hospital, Tokyo, Japan
| | - Masakazu Toi
- Department of Surgery (Breast Surgery), Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
22
|
Colomer R, Saura C, Sánchez-Rovira P, Pascual T, Rubio IT, Burgués O, Marcos L, Rodríguez CA, Martín M, Lluch A. Neoadjuvant Management of Early Breast Cancer: A Clinical and Investigational Position Statement. Oncologist 2019; 24:603-611. [PMID: 30710068 PMCID: PMC6516119 DOI: 10.1634/theoncologist.2018-0228] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Neoadjuvant treatment is increasingly one of the preferred therapeutic options for early breast cancer and may have some unique outcomes, such as identifying predictive and prognostic factors of response or increasing the knowledge of individual tumor biology. DESIGN A panel of experts from different specialties reviewed published clinical studies on the neoadjuvant management of breast cancer. Recommendations were made that emphasized the clinical multidisciplinary management and the investigational leverage in early breast cancer. RESULTS Neoadjuvant therapy has equivalent efficacy to adjuvant therapy, and it has some additional benefits that include increasing breast conservation, assessing tumor response, establishing prognosis based on the pathological response, and providing a "second opportunity" for nonresponding patients. Achieving pathological complete remission because of neoadjuvant therapy has been correlated with long-term clinical benefit, particularly in HER2-positive and triple-negative breast cancer. In addition, the neoadjuvant setting is a powerful model for the development of new drugs and the identification of prognostic markers. Finally, neoadjuvant therapy has proven to be cost-effective by reducing nondrug costs, avoiding radical surgery, and reducing hospital stays when compared with other treatment approaches. CONCLUSION Neoadjuvant therapy has clinical benefits in early breast cancer and provides in vivo information of individual breast cancer biology while allowing the investigation of new treatment approaches. Access to neoadjuvant therapy should be an option available to all patients with breast cancer through multidisciplinary tumor management. IMPLICATIONS FOR PRACTICE Neoadjuvant treatment should be strongly considered as a therapeutic option for localized breast cancer and is a powerful tool for understanding breast cancer biology and investigating new treatment approaches.
Collapse
Affiliation(s)
- Ramon Colomer
- Department of Medical Oncology, Hospital Universitario La Princesa, Madrid, Spain
| | - Cristina Saura
- Department of Medical Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | - Tomás Pascual
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Translational Genomic and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabel T Rubio
- Department of Breast Surgical Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Octavio Burgués
- Pathology Department, Hospital Clínico Universitario, Valencia, Spain
| | - Lourdes Marcos
- Department of Radiology, Hospital Universitario La Princesa, Madrid, Spain
| | - César A Rodríguez
- Department of Medical Oncology, Hospital Clínico Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Miguel Martín
- Department of Medical Oncology, Hospital Universitario Gregorio Marañon, Madrid, Spain
| | - Ana Lluch
- Department of Medical Oncology and Hematology, Hospital Clínico Universitario, University of Valencia-INCLIVA Health Research Institute, CIBERONC, Valencia, Spain
| |
Collapse
|
23
|
Post AEM, Bussink J, Sweep FCGJ, Span PN. Changes in DNA Damage Repair Gene Expression and Cell Cycle Gene Expression Do Not Explain Radioresistance in Tamoxifen-Resistant Breast Cancer. Oncol Res 2019; 28:33-40. [PMID: 31046897 PMCID: PMC7851527 DOI: 10.3727/096504019x15555794826018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tamoxifen-induced radioresistance, reported in vitro, might pose a problem for patients who receive neoadjuvant tamoxifen treatment and subsequently receive radiotherapy after surgery. Previous studies suggested that DNA damage repair or cell cycle genes are involved, and could therefore be targeted to preclude the occurrence of cross-resistance. We aimed to characterize the observed cross-resistance by investigating gene expression of DNA damage repair genes and cell cycle genes in estrogen receptor-positive MCF-7 breast cancer cells that were cultured to tamoxifen resistance. RNA sequencing was performed, and expression of genes characteristic for several DNA damage repair pathways was investigated, as well as expression of genes involved in different phases of the cell cycle. The association of differentially expressed genes with outcome after radiotherapy was assessed in silico in a large breast cancer cohort. None of the DNA damage repair pathways showed differential gene expression in tamoxifen-resistant cells compared to wild-type cells. Two DNA damage repair genes were more than two times upregulated (NEIL1 and EME2), and three DNA damage repair genes were more than two times downregulated (PCNA, BRIP1, and BARD1). However, these were not associated with outcome after radiotherapy in the TCGA breast cancer cohort. Genes involved in G1, G1/S, G2, and G2/M phases were lower expressed in tamoxifen-resistant cells compared to wild-type cells. Individual genes that were more than two times upregulated (MAPK13) or downregulated (E2F2, CKS2, GINS2, PCNA, MCM5, and EIF5A2) were not associated with response to radiotherapy in the patient cohort investigated. We assessed the expression of DNA damage repair genes and cell cycle genes in tamoxifen-resistant breast cancer cells. Though several genes in both pathways were differentially expressed, these could not explain the cross-resistance for irradiation in these cells, since no association to response to radiotherapy in the TCGA breast cancer cohort was found.
Collapse
Affiliation(s)
- Annemarie E M Post
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Johan Bussink
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Fred C G J Sweep
- Radboud University Medical Center, Department of Laboratory MedicineNijmegenThe Netherlands
| | - Paul N Span
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| |
Collapse
|
24
|
Selli C, Sims AH. Neoadjuvant Therapy for Breast Cancer as a Model for Translational Research. Breast Cancer (Auckl) 2019; 13:1178223419829072. [PMID: 30814840 PMCID: PMC6381436 DOI: 10.1177/1178223419829072] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 01/21/2023] Open
Abstract
Neoadjuvant therapy, where patients receive systemic therapy before surgical removal of the tumour, can downstage tumours allowing breast-conserving surgery, rather than mastectomy. In addition to its impact on surgery, the neoadjuvant setting offers a valuable opportunity to monitor individual tumour response. The effectiveness of standard and/or potential new therapies can be tested in the neoadjuvant pre-surgical setting. It can potentially help to identify markers differentiating patients that will potentially benefit from continuing with the same or a different adjuvant treatment enabling personalised treatment. Characterising the molecular response to treatment over time can more accurately identify the significant differences between baseline samples that would not be identified without post-treatment samples. In this review, we discuss the potential and challenges of using the neoadjuvant setting in translational breast cancer research, considering the implications for improving our understanding of response to treatment, predicting therapy benefit, modelling breast cancer dormancy, and the development of drug resistance.
Collapse
Affiliation(s)
- Cigdem Selli
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK
| |
Collapse
|
25
|
Guerrero-Zotano AL, Arteaga CL. Correction: Neoadjuvant Trials in ER+ Breast Cancer: A Tool for Acceleration of Drug Development and Discovery. Cancer Discov 2019; 9:304. [DOI: 10.1158/2159-8290.cd-18-1516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Dellapasqua S, Gray KP, Munzone E, Rubino D, Gianni L, Johansson H, Viale G, Ribi K, Bernhard J, Kammler R, Maibach R, Rabaglio-Poretti M, Ruepp B, Di Leo A, Coates AS, Gelber RD, Regan MM, Goldhirsch A, Colleoni M. Neoadjuvant Degarelix Versus Triptorelin in Premenopausal Patients Who Receive Letrozole for Locally Advanced Endocrine-Responsive Breast Cancer: A Randomized Phase II Trial. J Clin Oncol 2018; 37:386-395. [PMID: 30589600 DOI: 10.1200/jco.18.00296] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
PURPOSE To evaluate endocrine activity in terms of ovarian function suppression (OFS) of degarelix (a gonadotropin-releasing hormone [GnRH] antagonist) versus triptorelin (a GnRH agonist) in premenopausal patients receiving letrozole as neoadjuvant endocrine therapy for breast cancer. PATIENTS AND METHODS Premenopausal women with stage cT2 to 4b, any N, M0; estrogen receptor and progesterone receptor greater than 50%; human epidermal growth factor receptor 2-negative breast cancer were randomly assigned to triptorelin 3.75 mg administered intramuscularly on day 1 of every cycle or degarelix 240 mg administered subcutaneously (SC) on day 1 of cycle 1 then 80 mg SC on day 1 of cycles 2 through 6, both with letrozole 2.5 mg/day for six 28-day cycles. Surgery was performed 2 to 3 weeks after the last injection. Serum was collected at baseline, after 24 and 72 hours, at 7 and 14 days, and then before injections on cycles 2 through 6. The primary end point was time to optimal OFS (time from the first injection to first assessment of centrally assessed estradiol level ≤ 2.72 pg/mL [≤ 10 pmol/L] during neoadjuvant therapy). The trial had 90% power to detect a difference using a log-rank test with a two-sided α of .05. Secondary end points included response, tolerability, and patient-reported endocrine symptoms. RESULTS Between February 2014 and January 2017, 51 patients were enrolled (n = 26 received triptorelin plus letrozole; n = 25 received degarelix plus letrozole). Time to optimal OFS was three times faster for patients assigned to degarelix and letrozole than to triptorelin and letrozole (median, 3 v 14 days; hazard ratio, 3.05; 95% CI, 1.65 to 5.65; P < .001). Furthermore, OFS was maintained during subsequent cycles for all patients assigned to receive degarelix and letrozole, whereas 15.4% of patients assigned to receive triptorelin and letrozole had suboptimal OFS after cycle 1 (six events during 127 measurements). Adverse events as a result of both degarelix plus letrozole and triptorelin plus letrozole were as expected. CONCLUSION In premenopausal women receiving letrozole for neoadjuvant endocrine therapy, OFS was achieved more quickly and maintained more effectively with degarelix than with triptorelin.
Collapse
Affiliation(s)
- Silvia Dellapasqua
- 1 IEO, European Institute of Oncology Istituto di Recovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Kathryn P Gray
- 2 Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA
| | - Elisabetta Munzone
- 1 IEO, European Institute of Oncology Istituto di Recovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | | | - Lorenzo Gianni
- 4 Ospedale Infermi di Rimini, Azienda Unita Sanitaria (AUSL) della Romagna, Italy
| | - Harriet Johansson
- 1 IEO, European Institute of Oncology Istituto di Recovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giuseppe Viale
- 1 IEO, European Institute of Oncology Istituto di Recovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Karin Ribi
- 5 International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | - Jürg Bernhard
- 5 International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | - Roswitha Kammler
- 5 International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | - Rudolf Maibach
- 5 International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | | | - Barbara Ruepp
- 5 International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | - Angelo Di Leo
- 6 Hospital of Prato-AUSL Toscana Centro, Prato, Italy
| | - Alan S Coates
- 7 International Breast Cancer Study Group and University of Sydney, Sydney, Australia
| | - Richard D Gelber
- 8 Frontier Science and Technology Research Foundation and Harvard Medical School, Boston, MA.,9 International Breast Cancer Study Group Statistical Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Meredith M Regan
- 9 International Breast Cancer Study Group Statistical Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Aron Goldhirsch
- 1 IEO, European Institute of Oncology Istituto di Recovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Marco Colleoni
- 1 IEO, European Institute of Oncology Istituto di Recovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | | |
Collapse
|
27
|
Therapeutic predictors of neoadjuvant endocrine therapy response in estrogen receptor-positive breast cancer with reference to optimal gene expression profiling. Breast Cancer Res Treat 2018; 172:353-362. [PMID: 30151737 DOI: 10.1007/s10549-018-4933-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/21/2018] [Indexed: 12/27/2022]
Abstract
PURPOSE Neoadjuvant endocrine therapy (NAET) for estrogen receptor-positive primary breast cancer causes adequate tumor shrinkage, and is expected to be helpful for breast-conserving surgery, but the adaptation criteria, especially in regard to treatment duration, have never been elucidated. Re-visiting past gene expression profiles, we explored the data for specialized pre-therapeutic predictors and validated the results using our in-house clinical cohorts. METHODS We sorted the genes related to a > 30% tumor volume reduction through NAET from a cDNA microarray data-set of GSE20181, then selected the top 40 genes. We validated these gene expression levels using pre-therapeutic biopsy samples obtained from patients treated with long-term NAET (over 4 months; N = 40). A short-term (2-8 weeks; N = 37) NAET cohort was also validated to clarify whether expression of these genes is also related to a rapid response of Ki67 and PEPI score. RESULTS In the long-term group, higher expression of KRAS, CUL2, FAM13A, ADCK2, and LILRA2 was significantly associated with tumor shrinkage, and KRAS, MMS19, and IVD were related to lower PEPI score (≤ 3). Meanwhile in the short-term group, none of these genes except CUL2 showed a direct correlation with Ki67 reduction or PEPI score. This suggested that tumor shrinkage by NAET might be induced by response to the hypoxic environment (CUL2, FAM13A, KRAS) and activation of tumor immune system (LILRA2), without involving inhibition of proliferation. CONCLUSION Expression of specific genes may allow selection of the most responsive patients for maximum tumor shrinkage with NAET.
Collapse
|
28
|
Sun X, Ma X, Li Q, Yang Y, Xu X, Sun J, Yu M, Cao K, Yang L, Yang G, Zhang G, Wang X. Anti‑cancer effects of fisetin on mammary carcinoma cells via regulation of the PI3K/Akt/mTOR pathway: In vitro and in vivo studies. Int J Mol Med 2018; 42:811-820. [PMID: 29749427 PMCID: PMC6034928 DOI: 10.3892/ijmm.2018.3654] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 04/27/2018] [Indexed: 12/11/2022] Open
Abstract
Fisetin, a natural flavonoid found in a variety of edible and medical plants, has been suggested to inhibit the proliferation of various tumor cells and to induce apoptosis. However, the effects of fisetin on breast cancer have rarely been reported and the underlying mechanism is still undefined. The present study explored the anti‑cancer effects of fisetin on mammary carcinoma cells and the underlying mechanisms. Following treatment with fisetin, viability of 4T1, MCF‑7 and MDA‑MB‑231 cells were measured by MTT assay. The inhibitory effects of fisetin on proliferation, migration and invasion were evaluated in 4T1 cells using proliferation array, wound‑healing assay, and HUV‑EC‑C‑cell barrier based on electrical cell‑substrate impedance sensing platform. Cell apoptosis was analyzed by flow cytometry, and western blotting analysis was performed to identify target molecules. A 4T1 orthotopic mammary tumor model was used to assess the fisetin‑inhibition on tumor growth in vivo. Test kits were used to examine the liver and kidney function of tumor‑bearing mice. The results suggest that fisetin suppressed the proliferation of breast cancer cells, suppressed the metastasis and invasiveness of 4T1 cells, and induced the apoptosis of 4T1 cells in vitro. The potent anti‑cancer effect of fisetin was associated with the regulation of the phosphatidylinositol‑3‑kinase/protein kinase B/mammalian target of rapamycin pathway. In vivo experiments demonstrated that fisetin suppressed the growth of 4T1 cell‑derived orthotopic breast tumors and enhanced tumor cell apoptosis, and the evaluated alanine amino transferase and aspartate amino transferase levels in serum of tumor‑bearing mice suggested that fisetin may lead to side effects on liver biochemical function. The present study confirms that fisetin exerted an anti‑mammary carcinoma effect. However, in vivo experiments also revealed that fisetin had low solubility and low bioavailability. Further investigation is required to determine the clinical value of fisetin.
Collapse
Affiliation(s)
- Xu Sun
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
| | - Xueman Ma
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
- Department of Oncology, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120
| | - Qiwei Li
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029
| | - Yong Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029
| | - Xiaolong Xu
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
- Department of Immunology, Beijing Institute of Traditional Chinese Medicine, Beijing 100010
| | - Jiaqi Sun
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
- Department of Gynecology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, P.R. China
| | - Mingwei Yu
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
| | - Kexin Cao
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
| | - Lin Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
| | - Guowang Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
| | - Xiaomin Wang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010
| |
Collapse
|
29
|
Post AEM, Smid M, Nagelkerke A, Martens JWM, Bussink J, Sweep FCGJ, Span PN. Interferon-Stimulated Genes Are Involved in Cross-resistance to Radiotherapy in Tamoxifen-Resistant Breast Cancer. Clin Cancer Res 2018; 24:3397-3408. [PMID: 29661777 DOI: 10.1158/1078-0432.ccr-17-2551] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/07/2018] [Accepted: 04/10/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Treatment resistance is the main cause of adverse disease outcome in breast cancer patients. Here, we aimed to investigate common features in tamoxifen-resistant and radioresistant breast cancer, as tamoxifen-resistant breast cancer cells are cross-resistant to irradiation in vitroExperimental Design: RNA sequencing of tamoxifen-resistant and radioresistant breast cancer cells was performed and validated by quantitative PCR. Pathways were further investigated in vitro and in breast cancer patient cohorts to establish their relation with treatment resistance.Results: Both tamoxifen-resistant and radioresistant breast cancer cells had increased expression levels of genes involved in type I IFN signaling compared with nonresistant cells. IFN-stimulated genes (ISG) were induced in a dose-dependent and time-dependent manner after tamoxifen treatment and irradiation. Tamoxifen treatment also led to ssDNA presence in the cytoplasm, which is known to induce expression of ISGs, a phenomenon that has already been described for irradiation. Moreover, in a breast cancer patient cohort, high expression levels of ISGs were found in the primary tumor in around half of the patients. This was associated with a tumor-infiltrating lymphocyte (TIL) expression signature, although the ISGs were also expressed by the tumor cells themselves. Importantly, the expression of ISGs correlated with outcome in breast cancer patients treated with adjuvant tamoxifen or radiotherapy, but not in systemically untreated patients or chemotherapy-treated patients.Conclusions: Our data indicate that expression of ISGs by tumor cells is involved in acquired, treatment-induced resistance to tamoxifen and radiotherapy, and might play a role in intrinsic resistance via interaction with TILs. Clin Cancer Res; 24(14); 3397-408. ©2018 AACR.
Collapse
Affiliation(s)
- Annemarie E M Post
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands. .,Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Marcel Smid
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Anika Nagelkerke
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands.,Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Paul N Span
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
30
|
Reinert T, Gonçalves R, Ellis MJ. Current Status of Neoadjuvant Endocrine Therapy in Early Stage Breast Cancer. Curr Treat Options Oncol 2018; 19:23. [PMID: 29663173 DOI: 10.1007/s11864-018-0538-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OPINION STATEMENT Neoadjuvant endocrine therapy (NET) with Ki67-based response monitoring is a practical, cost-effective approach to the management of clinical stage II and III estrogen receptor-positive (ER+) breast cancer. In addition to marked improvements in rates of breast conservation, the identification of extreme responders on the basis of the preoperative endocrine prognostic index (PEPI) provides a rationale to avoid chemotherapy on the basis of highly favorable prognosis in some patients. Finally, samples accrued from patients treated with neoadjuvant therapy are providing valuable insights into the molecular basis for intrinsic resistance to endocrine therapy and promise a more rational basis and precise approach to the systemic treatment of ER+ breast cancer.
Collapse
Affiliation(s)
- Tomás Reinert
- Hospital do Câncer Mãe de Deus, Porto Alegre, Brazil
- Postgraduation Department of Medical Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rodrigo Gonçalves
- Setor de Mastologia, Hospital das Clínicas, Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Matthew J Ellis
- Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|