51
|
Kong J, Zhu S, Shi G, Liu Z, Zhang J, Ren J. Prediction of Locoregional Recurrence-Free Survival of Oesophageal Squamous Cell Carcinoma After Chemoradiotherapy Based on an Enhanced CT-Based Radiomics Model. Front Oncol 2021; 11:739933. [PMID: 34631575 PMCID: PMC8499696 DOI: 10.3389/fonc.2021.739933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Chemoradiotherapy is the standard treatment for moderate and advanced oesophageal cancer. The aim of this study was to establish a predictive model based on enhanced computed tomography examination, and to evaluate its clinical value for detecting locoregional recurrence-free survival (LRFS) in cases of oesophageal squamous cell carcinoma after radiotherapy. MATERIALS AND METHODS In total, 218 patients with pathologically diagnosed oesophageal squamous cell carcinoma who received radical chemoradiotherapy from July 2016 to December 2017 were collected in this study. Patients were randomly divided into either a training group (n=153) or a validation group (n=65) in a 7:3 ratio. Clinical patient information was then recorded. The enhanced computed tomography scan images of the patients were imported into 3D-slicer software (version 4.8.1), and the radiomic features were extracted by the Python programme package. In the training group, the dimensionality reduction of the radiomic features was implemented by Lasso regression, and then a radiological label, the model of predicting LRFS, was established and evaluated. To achieve a better prediction performance, the radiological label was combined with clinical risk factor information to construct a radiomics nomogram. A receiver operating characteristic curve was used to evaluate the efficacy of different models. Calibration curves were used to assess the consistency between the predicted and observed recurrence risk, and the Hosmer-Lemeshow method was used to test model fitness. The C-index evaluated the discriminating ability of the prediction model. Decision curve analysis was used to determine the clinical value of the constructed prediction model. RESULTS Of the 218 patients followed up in this study, 44 patients (28.8%) in the training group and 21 patients (32.3%) in the validation group experienced recurrence. There was no difference in LRFS between the two groups (χ2 = 0.525, P=0.405). Lasso regression was used in the training group to select six significant radiomic features. The radiological label established using these six features had a satisfactory prediction performance. The C-index was 0.716 (95% CI: 0.645-0.787) in the training group and 0.718 (95% CI: 0.612-0.825) in the validation group. The radiomics nomogram, which included the radiological label and clinical risk factors, achieved a better prediction than the radiological label alone. The C-index was 0.742 (95% CI: 0.674-0.810) in the training group and 0.715 (95% CI: 0.609-0.820) in the validation group. The results of the calibration curve and decision curve analyses indicated that the radiomics nomogram was superior in predicting LRFS of oesophageal carcinoma after radiotherapy. CONCLUSIONS A radiological label was successfully established to predict the LRFS of oesophageal squamous cell carcinoma after radiotherapy. The radiomics nomogram was complementary to the clinical prognostic features and could improve the prediction of the LRFS after radiotherapy for oesophageal cancer.
Collapse
Affiliation(s)
- Jie Kong
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuchai Zhu
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gaofeng Shi
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhikun Liu
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Zhang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jialiang Ren
- Pharmaceutical Diagnosis, GE Healthcare, Beijing, China
| |
Collapse
|
52
|
Zhou Q, Qian Z, Ding W, Jiang G, Sun C, Xu K. Chronic Psychological Stress Attenuates the Efficacy of anti-PD-L1 Immunotherapy for Bladder Cancer in Immunocompetent Mice. Cancer Invest 2021; 39:571-581. [PMID: 34148483 DOI: 10.1080/07357907.2021.1943746] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We aimed to explore whether chronic psychological stress affects the efficacy of immune checkpoint inhibitors (ICIs) immunotherapy in bladder cancer. The chronic unpredictable mild stress (CUMS) process was applied during the administration of anti-PD-L1 for subcutaneous tumors in mice. Tumor regression was obviously shown in anti-PD-L1 therapy groups, while this effect was notably attenuated by CUMS. Additionally, increased infiltration of regulatory T-cells, decreased amount of CD8+ lymphocytes, and reduced levels of tumor-associated cytokines in tumor sites were observed in mice treated with anti-PD-L1 under CUMS. Therefore, chronic psychological stress could weaken the potency of anti-PD-L1 immunotherapy for bladder cancer.
Collapse
Affiliation(s)
- Qidong Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiyu Qian
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Guangliang Jiang
- Department of Urology, Ruijin Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Chuanyu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
53
|
Wu C, Jiang H, Chen J. A Systematic Review and Meta-Analysis about the Effect of Bisphosphonates on the Risk of Skeletal-Related Event in Men with Prostate Cancer. Anticancer Agents Med Chem 2021; 20:1604-1612. [PMID: 32436834 DOI: 10.2174/1871520620666200521114815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/29/2020] [Accepted: 02/10/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND Although the adjuvant therapy of bisphosphonates in prostate cancer is effective in improving bone mineral density, it is still uncertain whether bisphosphonates could decrease the risk of Skeletal- Related Event (SRE) in patients with prostate cancer. We reviewed and analyzed the effect of different types of bisphosphonates on the risk of SRE, defined as pathological fracture, spinal cord compression, radiation therapy to the bone, surgery to bone, hypercalcemia, bone pain, or death as a result of prostate cancer. METHODS A systemic literature search was conducted on PubMed and related bibliographies. The emphasis during data extraction was laid on the Hazard Ratio (HR) and the corresponding 95% Confidence Interval (CI) from every eligible Randomized Controlled Trial (RCT). HR was pooled with the fixed effects model, and preplanned subgroup analyses were performed. RESULTS 5 RCTs (n = 4651) were included and analyzed finally after screening 51 articles. The meta-analysis of all participants showed no significant decrease in the risk of SRE when adding bisphosphonates to control group (HR = 0.968, 95% CI = 0.874 - 1.072, p = 0.536) with low heterogeneity (I2 = 0.0% (d.f. = 4) p = 0.679). There was no significant improvement on SRE neither in the subgroups with Metastases (M1) or Castration-Sensitive Prostate Cancer (CSPC) (respectively HR = 0.968, 95% CI = 0.874 - 1.072, p = 0.536, I2 = 0.0% (d.f. = 4) p = 0.679; HR = 0.954, 95% CI = 0.837 - 1.088, p = 0.484, I2 = 0.0% (d.f. = 3) p = 0.534). CONCLUSION Our study demonstrated that bisphosphonates could not statistically significantly reduce the risk of SRE in patients with prostate cancer, neither in the subgroups with M1 or CSPC.
Collapse
Affiliation(s)
- Congcong Wu
- Department of Chemotherapy and Radiotherapy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang Province, China
| | - Hua Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
54
|
Hu R, Tao T, Yu L, Ding Q, Zhu G, Peng G, Zheng S, Yang L, Wu S. Multi-Omics Characterization of Tumor Microenvironment Heterogeneity and Immunotherapy Resistance Through Cell States-Based Subtyping in Bladder Cancer. Front Cell Dev Biol 2021; 9:809588. [PMID: 35223867 PMCID: PMC8864284 DOI: 10.3389/fcell.2021.809588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023] Open
Abstract
Due to the strong heterogeneity of bladder cancer (BC), there is often substantial variation in the prognosis and efficiency of immunotherapy among BC patients. For the precision treatment and assessment of prognosis, the subtyping of BC plays a critical role. Despite various subtyping methods proposed previously, most of them are based on a limited number of molecules, and none of them is developed on the basis of cell states. In this study, we construct a single-cell atlas by integrating single cell RNA-seq, RNA microarray, and bulk RNA-seq data to identify the absolute proportion of 22 different cell states in BC, including immune and nonimmune cell states derived from tumor tissues. To explore the heterogeneity of BC, BC was identified into four different subtypes in multiple cohorts using an improved consensus clustering algorithm based on cell states. Among the four subtypes, C1 had median prognosis and best overall response rate (ORR), which characterized an immunosuppressive tumor microenvironment. C2 was enriched in epithelial-mesenchymal transition/invasion, angiogenesis, immunosuppression, and immune exhaustion. Surely, C2 performed the worst in prognosis and ORR. C3 with worse ORR than C2 was enriched in angiogenesis and almost nonimmune exhaustion. Displaying an immune effective environment, C4 performed the best in prognosis and ORR. We found that patients with just an immunosuppressive environment are suitable for immunotherapy, but patients with an immunosuppressive environment accompanied by immune exhaustion or angiogenesis may resist immunotherapy. Furthermore, we conducted exploration into the heterogeneity of the transcriptome, mutational profiles, and somatic copy-number alterations in four subtypes, which could explain the significant differences related to cell states in prognosis and ORR. We also found that PD-1 in immune and tumor cells could both influence ORR in BC. The level of TGFβ in a cell state can be opposite to the overall level in the tissues, and the level in a specific cell state could predict ORR more accurately. Thus, our work furthers the understanding of heterogeneity and immunotherapy resistance in BC, which is expected to assist clinical practice and serve as a supplement to the current subtyping method from a novel perspective of cell states.
Collapse
Affiliation(s)
- Rixin Hu
- Health Science Center, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Tao Tao
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Lu Yu
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
- Teaching Center of Shenzhen Luohu Hospital, Shantou University Medical College, Shantou, China
| | - Qiuxia Ding
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Guanghui Zhu
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Guoyu Peng
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Shiwen Zheng
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Leyun Yang
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
| | - Song Wu
- Health Science Center, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
- Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, China
- Teaching Center of Shenzhen Luohu Hospital, Shantou University Medical College, Shantou, China
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
- *Correspondence: Song Wu,
| |
Collapse
|
55
|
Perry LM, Bateni SB, Sauder CAM. ASO Author Reflections: Deescalating Therapy for Older Men with Early Estrogen Receptor Positive Breast Cancer. Ann Surg Oncol 2020; 28:2472-2473. [PMID: 33169302 DOI: 10.1245/s10434-020-09357-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 10/28/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Lauren M Perry
- Division of Surgical Oncology, Department of Surgery, University of California, Davis Medical Center, UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Sarah B Bateni
- Division of Surgical Oncology, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Candice A M Sauder
- Division of Surgical Oncology, Department of Surgery, University of California, Davis Medical Center, UC Davis Comprehensive Cancer Center, Sacramento, CA, USA.
| |
Collapse
|
56
|
Bradley SA, Zhang J, Jensen MK. Deploying Microbial Synthesis for Halogenating and Diversifying Medicinal Alkaloid Scaffolds. Front Bioeng Biotechnol 2020; 8:594126. [PMID: 33195162 PMCID: PMC7644825 DOI: 10.3389/fbioe.2020.594126] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/02/2020] [Indexed: 11/13/2022] Open
Abstract
Plants produce some of the most potent therapeutics and have been used for thousands of years to treat human diseases. Today, many medicinal natural products are still extracted from source plants at scale as their complexity precludes total synthesis from bulk chemicals. However, extraction from plants can be an unreliable and low-yielding source for human therapeutics, making the supply chain for some of these life-saving medicines expensive and unstable. There has therefore been significant interest in refactoring these plant pathways in genetically tractable microbes, which grow more reliably and where the plant pathways can be more easily engineered to improve the titer, rate and yield of medicinal natural products. In addition, refactoring plant biosynthetic pathways in microbes also offers the possibility to explore new-to-nature chemistry more systematically, and thereby help expand the chemical space that can be probed for drugs as well as enable the study of pharmacological properties of such new-to-nature chemistry. This perspective will review the recent progress toward heterologous production of plant medicinal alkaloids in microbial systems. In particular, we focus on the refactoring of halogenated alkaloids in yeast, which has created an unprecedented opportunity for biosynthesis of previously inaccessible new-to-nature variants of the natural alkaloid scaffolds.
Collapse
Affiliation(s)
| | | | - Michael K. Jensen
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
57
|
Bateni SB, Perry LM, Zhao X, Arora M, Daly ME, Stewart SL, Bold RJ, Canter RJ, Sauder CAM. The Role of Radiation Therapy in Addition to Lumpectomy and Hormone Therapy in Men 70 Years of Age and Older with Early Breast Cancer: A NCDB Analysis. Ann Surg Oncol 2020; 28:2463-2471. [PMID: 33094375 DOI: 10.1245/s10434-020-09242-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/03/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Current treatment guidelines for male breast cancer are guided by female-only trials despite data suggesting distinct clinicopathologic differences between sexes. We sought to evaluate whether radiation therapy (RT) after lumpectomy was associated with equivalent survival among men > 70 years of age with stage I, estrogen receptor (ER) positive tumors, as seen in women from the Cancer and Leukemia Group B (CALGB) 9343 trial. METHODS We performed a retrospective analysis of 752 stage I, ER-positive male breast cancer patients ≥ 70 years who were treated with hormone therapy and surgery, with or without RT, from the National Cancer Database between 2004 and 2014. Patients were categorized based on surgery and RT (lumpectomy alone, lumpectomy with RT, and mastectomy alone). Multivariable Cox proportional hazards regression analysis was used to compare overall survival between treatment groups. RESULTS Most patients underwent total mastectomy, with only 32.6% treated with lumpectomy. Of those who underwent lumpectomy, 72.7% received adjuvant RT. In multivariate analysis, there was no statistical difference in overall survival when comparing lumpectomy alone and lumpectomy with RT (aHR 0.72 [95% CI 0.38-1.37], p = 0.31) or when comparing lumpectomy (alone or with RT) and mastectomy (aHR 1.28 [95% CI 0.88-1.87], p = 0.20). CONCLUSIONS In this national sample of elderly men with ER-positive early-stage disease treated with endocrine therapy, there were no significant differences in overall survival when comparing lumpectomy alone and lumpectomy with RT, or lumpectomy (alone or with RT) and mastectomy. These results suggest that less aggressive treatment may be appropriate for a subset of male breast cancer patients.
Collapse
Affiliation(s)
- Sarah B Bateni
- Division of Surgical Oncology, Department of Surgery, Davis Medical Center, UC Davis Comprehensive Cancer Center, University of California, Sacramento, CA, USA
| | - Lauren M Perry
- Division of Surgical Oncology, Department of Surgery, Davis Medical Center, UC Davis Comprehensive Cancer Center, University of California, Sacramento, CA, USA
| | - Xiao Zhao
- Department of Radiation Oncology, Davis Medical Center, University of California, Sacramento, CA, USA
| | - Mili Arora
- Division of Hematology and Oncology, Department of Internal Medicine, Davis Medical Center, University of California, Sacramento, CA, USA
| | - Megan E Daly
- Department of Radiation Oncology, Davis Medical Center, University of California, Sacramento, CA, USA
| | - Susan L Stewart
- Department of Public Health Sciences, Davis Medical Center, University of California, Sacramento, CA, USA
| | - Richard J Bold
- Division of Surgical Oncology, Department of Surgery, Davis Medical Center, UC Davis Comprehensive Cancer Center, University of California, Sacramento, CA, USA
| | - Robert J Canter
- Division of Surgical Oncology, Department of Surgery, Davis Medical Center, UC Davis Comprehensive Cancer Center, University of California, Sacramento, CA, USA
| | - Candice A M Sauder
- Division of Surgical Oncology, Department of Surgery, Davis Medical Center, UC Davis Comprehensive Cancer Center, University of California, Sacramento, CA, USA.
| |
Collapse
|
58
|
Johdi NA, Sukor NF. Colorectal Cancer Immunotherapy: Options and Strategies. Front Immunol 2020; 11:1624. [PMID: 33042104 PMCID: PMC7530194 DOI: 10.3389/fimmu.2020.01624] [Citation(s) in RCA: 287] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is the third most common cancer in the world with increasing incidence and mortality rates globally. Standard treatments for colorectal cancer have always been surgery, chemotherapy and radiotherapy which may be used in combination to treat patients. However, these treatments have many side effects due to their non-specificity and cytotoxicity toward any cells including normal cells that are growing and dividing. Furthermore, many patients succumb to relapse even after a series of treatments. Thus, it is crucial to have more alternative and effective treatments to treat CRC patients. Immunotherapy is one of the new alternatives in cancer treatment. The strategy is to utilize patients' own immune systems in combating the cancer cells. Cancer immunotherapy overcomes the issue of specificity which is the major problem in chemotherapy and radiotherapy. The normal cells with no cancer antigens are not affected. The outcomes of some cancer immunotherapy have been astonishing in some cases, but some which rely on the status of patients' own immune systems are not. Those patients who responded well to cancer immunotherapy have a better prognostic and better quality of life.
Collapse
Affiliation(s)
- Nor Adzimah Johdi
- UKM Medical Molecular Biology Institute (UMBI), National University of Malaysia, Bangi, Malaysia
| | | |
Collapse
|
59
|
Vengaloor Thomas T, Gordy XZ, Lirette ST, Albert AA, Gordy DP, Vijayakumar S, Vijayakumar V. Lack of Racial Survival Differences in Metastatic Prostate Cancer in National Cancer Data Base (NCDB): A Different Finding Compared to Non-metastatic Disease. Front Oncol 2020; 10:533070. [PMID: 33072567 PMCID: PMC7531281 DOI: 10.3389/fonc.2020.533070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Inconsistent findings have been reported in the literature regarding racial differences in survival outcomes between African American and white patients with metastatic prostate cancer (mPCa). The current study utilized a national database to determine whether racial differences exist among the target population to address this inconsistency. Methods: This study retrospectively reviewed prostate cancer (PCa) patient data (N = 1,319,225) from the National Cancer Database (NCDB). The data were divided into three groupings based on the metastatic status: (1) no metastasis (N = 318,291), (2) bone metastasis (N = 29,639), and (3) metastases to locations other than bone, such as brain, liver, or lung (N = 952). Survival probabilities of African American and white PCa patients with bone metastasis were examined through parametric proportional hazards Weibull models and Bayesian survival analysis. These results were compared to patients with no metastasis or other types of metastases. Results: No statistically supported racial disparities were observed for African American and white men with bone metastasis (p = 0.885). Similarly, there were no racial disparities in survival for those men suffering from other metastases (liver, lung, or brain). However, racial disparities in survival were observed among the two racial groups with non-metastatic PCa (p < 0.001) or when metastasis status was not taken into account (p < 0.001). The Bayesian analysis corroborates the finding. Conclusion: This research supports our previous findings and shows that there are no racial differences in survival outcomes between African American and white patients with mPCa. In contrast, racial disparities in the survival outcome continue to exist among non-metastatic PCa patients. Further research is warranted to explain this difference.
Collapse
Affiliation(s)
- Toms Vengaloor Thomas
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Xiaoshan Z Gordy
- Department of Health Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Seth T Lirette
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ashley A Albert
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, United States
| | - David P Gordy
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Srinivasan Vijayakumar
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Vani Vijayakumar
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
60
|
Dermatologic adverse events related to the PI3Kα inhibitor alpelisib (BYL719) in patients with breast cancer. Breast Cancer Res Treat 2020; 183:227-237. [PMID: 32613539 DOI: 10.1007/s10549-020-05726-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Rash develops in approximately 50% of patients receiving alpelisib for breast cancer, often requiring dose modifications. Here, we describe the clinicopathologic, laboratory, and management characteristics of alpelisib-related dermatologic adverse events (dAEs). METHODS A single center-retrospective analysis was conducted. Data were abstracted from electronic medical records. RESULTS A total of 102 patients (mean age 56 years, range 27-83) receiving alpelisib most frequently in combination with endocrine therapy (79, 77.5%) were included. We identified 41 (40.2%) patients with all-grade rash distributed primarily along the trunk (78%) and extremities (70%) that developed approximately within two weeks of treatment initiation (mean 12.8 ± 1.5 days) and lasted one-week (mean duration 7.1 ± 0.8 days). Of 29 patients with documented morphology of alpelisib-related dAEs, 26 (89.7%) had maculopapular rash. Histology showed perivascular and interface lymphocytic dermatitis. All-grade rash correlated with an increase in serum eosinophils from 2.7 to 4.4%, p < 0.05, and prophylaxis with non-sedating antihistamines (n = 43) was correlated with a reduction of grade 1/2 rash (OR 0.39, p = 0.09). Sixteen (84.2%) of 19 patients with grade 3 dAEs resulted in interruption of alpelisib, which were managed with antihistamines, topical and systemic corticosteroids. We did not observe rash recurrence in 12 (75%) patients who were re-challenged. CONCLUSIONS A maculopapular rash associated with increased blood eosinophils occurs frequently with alpelisib. While grade 3 rash leads to alpelisib therapy interruption, dermatologic improvement is evident with systemic corticosteroids; and most patients can continue oncologic treatment at a maintained or reduced dose upon re-challenge with alpelisib.
Collapse
|
61
|
The five “W”s and “How” of Targeted Alpha Therapy: Why? Who? What? Where? When? and How? RENDICONTI LINCEI-SCIENZE FISICHE E NATURALI 2020. [DOI: 10.1007/s12210-020-00900-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
62
|
Wong KY, Tan AMN. Short term outcomes of minimally invasive selective lateral pelvic lymph node dissection for low rectal cancer. World J Gastrointest Surg 2020; 12:178-189. [PMID: 32426097 PMCID: PMC7215974 DOI: 10.4240/wjgs.v12.i4.178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pelvic recurrence after rectal cancer surgery is still a significant problem despite the introduction of total mesorectal excision and chemoradiation treatment (CRT), and one of the most common areas of recurrence is in the lateral pelvic lymph nodes. Hence, there is a possible role for lateral pelvic lymph node dissection (LPND) in rectal cancer.
AIM To evaluate the short-term outcomes of patients who underwent minimally invasive LPND during rectal cancer surgery. Secondary outcomes were to evaluate for any predictive factors to determine lymph node metastases based on pre-operative scans.
METHODS From October 2016 to November 2019, 22 patients with stage II or III rectal cancer underwent minimally invasive rectal cancer surgery and LPND. These patients were all discussed at a multidisciplinary tumor board meeting and most of them received neoadjuvant chemoradiation prior to surgery. All patients had radiologically positive lateral pelvic lymph nodes on the initial staging scans, defined as lymph nodes larger than 7 mm in long axis measurement, or abnormal radiological morphology. LPND was only performed on the involved side.
RESULTS Majority of the patients were male (18/22, 81.8%), with a median age of 65 years (44-81). Eighteen patients completed neoadjuvant CRT pre-operatively. 18 patients (81.8%) had unilateral LPND, with the others receiving bilateral surgery. The median number of lateral pelvic lymph nodes harvested was 10 (3-22) per pelvic side wall. 8 patients (36.4%) had positive metastases identified in the lymph nodes harvested. The median pre-CRT size of these positive lymph nodes was 10mm. Median length of stay was 7.5 d (3-76), and only 2 patients failed initial removal of their urinary catheter. Complication rates were low, with only 1 lymphocele and 1 anastomotic leak. There was only 1 mortality (4.5%). There have been no recurrences so far.
CONCLUSION Chemoradiation is inadequate in completely eradicating lateral wall metastasis and there are still technical limitations in accurately diagnosing metastases in these areas. A pre-CRT lymph node size of ≥ 10 mm is suggestive of metastases. LPND may be performed safely with minimally invasive surgery.
Collapse
Affiliation(s)
- Kar Yong Wong
- Colorectal Surgery Service, Department of General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Aloysius MN Tan
- Colorectal Surgery Service, Department of General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
| |
Collapse
|
63
|
Gal9/Tim-3 expression level is higher in AML patients who fail chemotherapy. J Immunother Cancer 2019; 7:175. [PMID: 31291985 PMCID: PMC6621946 DOI: 10.1186/s40425-019-0611-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/06/2019] [Indexed: 01/07/2023] Open
Abstract
Immune checkpoint pathways active in Acute Myeloid Leukemia (AML) patients, especially during the course of remission induction chemotherapy, have not been well studied. Although dominant in mediating T cell dysfunction in cancer, it is now well-accepted that interruption of PD-1/PD-L1 axes alone does not always completely restore T cell function, indicating the involvement of additional negative regulatory pathways, such as TIM-3/Gal-9, in promoting T cell exhaustion.Here, we characterized these pathways in AML patients enrolled in a phase I dose escalation trial that combined Selinexor, a Selective Inhibitor of Nuclear Export (SINE), with high-dose cytarabine (HiDAC) and mitoxantrone (Mito) (NCT02573363) as induction therapy.To monitor changes in expression of immune checkpoint receptors, multi-parameter flow cytometry was performed on peripheral blood and bone marrow biopsy specimens at diagnosis and following induction therapy in 26 AML patients. Expression of CD47, PD-L1, PD-L2 and Gal9 was assessed on CD34+ AML blasts, as well as on CD34- cell populations. In parallel, we evaluated expression of inhibitory (PD1, CTLA4, LAG3, TIM-3) and stimulatory (CD28, ICOS, CD137, OX40, CD40L, HLA-DR) co-receptors on CD4+ and CD8+ T cell subsets.Compared to baseline, the frequency of Gal9+ CD34- cells was significantly higher in patients with treatment failure (TF) than in those in complete remission (CR), and this finding correlated with increased TIM-3 expression on marrow-resident T cells in TF patients. Moreover, when we measured the expression level of PD-1 and TIM-3 in bone marrow samples compared to peripheral blood, TIM-3 was significantly higher in BM specimens.Our results suggest that targeting the Gal9/Tim-3 axis could be effective in combination with induction chemotherapy to increase the likelihood of complete remission in AML patients.
Collapse
|
64
|
Tumour necrosis as assessed with 18F-FDG PET is a potential prognostic marker in diffuse large B cell lymphoma independent of MYC rearrangements. Eur Radiol 2019; 29:6018-6028. [PMID: 31028445 PMCID: PMC6795618 DOI: 10.1007/s00330-019-06178-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 12/14/2022]
Abstract
Objectives MYC gene rearrangements in diffuse large B cell lymphomas (DLBCLs) result in high proliferation rates and are associated with a poor prognosis. Strong proliferation is associated with high metabolic demand and tumour necrosis. The aim of this study was to investigate differences in the presence of necrosis and semiquantitative 18F-FDG PET metrics between DLBCL cases with or without a MYC rearrangement. The prognostic impact of necrosis and semiquantitative 18F-FDG PET parameters was investigated in an explorative survival analysis. Methods Fluorescence in situ hybridisation analysis for MYC rearrangements, visual assesment, semiquantitative analysis of 18F-FDG PET scans and patient survival analysis were performed in 61 DLBCL patients, treated at a single referral hospital between 2008 and 2015. Results Of 61 tumours, 21 (34%) had a MYC rearrangement (MYC+). MYC status was neither associated with the presence of necrosis on 18F-FDG PET scans (necrosisPET; p = 1.0) nor associated with the investigated semiquantitative parameters maximum standard uptake value (SUVmax; p = 0.43), single highest SUVmax (p = 0.49), metabolic active tumour volume (MATV; p = 0.68) or total lesion glycolysis (TLG; p = 0.62). A multivariate patient survival analysis of the entire cohort showed necrosisPET as an independent prognostic marker for disease-specific survival (DSS) (HR = 13.9; 95% CI 3.0–65; p = 0.001). Conclusions MYC rearrangements in DLBCL have no influence on the visual parameter necrosisPET or the semi-quantiative parameters SUVmax, MATV and TLG. Irrespective of MYC rearrangements, necrosisPET is an independent, adverse prognostic factor for DSS. Key Points • Retrospective analysis indicates that MYC rearrangement is not associated with necrosis on18F-FDG PET (necrosisPET) scans or semiquantitative18F-FDG PET parameters. • NecrosisPETis a potential independent adverse prognostic factor for disease-specific survival in patients with DLBCL and is not influenced by the presence of MYC rearrangements. Electronic supplementary material The online version of this article (10.1007/s00330-019-06178-9) contains supplementary material, which is available to authorized users.
Collapse
|
65
|
Sowder ME, Johnson RW. Bone as a Preferential Site for Metastasis. JBMR Plus 2019; 3:e10126. [PMID: 30918918 PMCID: PMC6419612 DOI: 10.1002/jbm4.10126] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 10/28/2018] [Indexed: 02/06/2023] Open
Abstract
Bone marrow provides a unique microenvironment favoring the colonization and outgrowth of metastatic tumor cells. Despite the high incidence of bone metastasis in breast and prostate cancer patients, many of the molecular mechanisms controlling metastatic progression remain unclear. Several gene signatures associated with bone metastasis have been reported, but no metastasis-specific gene alterations have been identified. Therefore, there has been considerable interest in understanding how the bone microenvironment impacts the behavior of disseminated tumor cells (DTCs) prior to and following colonization of the bone. Substantial evidence indicates that disruption of normal bone homeostasis by tumor-derived factors establishes a premetastatic niche within the bone that favors DTC colonization. Following dissemination, bone resident cells and the surrounding stroma provide critical signals that support tumor cell colonization, survival, and eventual outgrowth. Clinical data suggest that patients can harbor DTCs for years to decades prior to developing overt bone metastases, suggesting a period of tumor dormancy occurs in the bone marrow. Several dormancy-promoting factors have been recently identified; however, critical questions surrounding the molecular triggers and timing of tumor cell emergence from dormancy remain. Here, we review how metastatic tumor cells co-opt the bone marrow microenvironment for metastatic progression and discuss emerging insights into how to more effectively target DTCs and prevent metastasis. © 2018 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Miranda E Sowder
- Program in Cancer BiologyVanderbilt UniversityNashvilleTNUSA
- Vanderbilt Center for Bone BiologyDepartment of MedicineDivision of Clinical PharmacologyVanderbilt University Medical CenterNashvilleTNUSA
| | - Rachelle W Johnson
- Program in Cancer BiologyVanderbilt UniversityNashvilleTNUSA
- Vanderbilt Center for Bone BiologyDepartment of MedicineDivision of Clinical PharmacologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of MedicineDivision of Clinical PharmacologyVanderbilt University Medical CenterNashvilleTNUSA
| |
Collapse
|
66
|
Wang SQ, Cui SX, Qu XJ. Metformin inhibited colitis and colitis-associated cancer (CAC) through protecting mitochondrial structures of colorectal epithelial cells in mice. Cancer Biol Ther 2018; 20:338-348. [PMID: 30359174 DOI: 10.1080/15384047.2018.1529108] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although a mountain of papers have showed that metformin plays a role in inhibiting cancers, but the mechanism underpinning this has not yet fully elucidated. Herein, we used AOM/DSS model, the clinicopathological features are similar to those found in humans, to investigate the effects of metformin as well as combination with 5-FU in the prevention of colitis and colitis associated cancer (CAC). Oral metformin significantly inhibited DSS-induced ulcerative colitis and AOM/DSS-induced CAC. Metformin also ameliorated 5-FU-induced colorectal gastrointestinal symptoms in mice. Metformin combination with 5-FU strongly inhibited colorectal cancer. Metformin reduced levels of the NFκB signaling components p-IKKα/β, p-NFκB, p-IκBα in colorectal mucosal cells. Transmission electron microscopy analysis suggested that the inhibition of metformin on colitis and CAC might associate with its biological activity of protecting mitochondrial structures of colorectal epithelial cells. Further analysis by Mito Tracker Red staining assay indicated that metformin prevented H2O2-induced mitochondrial fission correlated with a decrease of mitochondrial perimeter. In addition, metformin increased the level of NDUFA9, a Q-module subunit required for complex I assembly, in colorectal epithelial cells. These observations of metformin in the inhibition of colitis and CAC might associate with its activity of activating the LKB1/AMPK pathway in colorectal epithelial cells. In conclusion, metformin inhibited colitis and CAC through protecting the mitochondrial structures of colorectal epithelial cells.
Collapse
Affiliation(s)
- Shu-Qing Wang
- a Department of Pharmacology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Shu-Xiang Cui
- b Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , China
| | - Xian-Jun Qu
- a Department of Pharmacology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| |
Collapse
|
67
|
Cardiotoxicity of trastuzumab in patients with HER2-positive gastric cancer. Oncotarget 2017; 8:61837-61845. [PMID: 28977908 PMCID: PMC5617468 DOI: 10.18632/oncotarget.18700] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 05/15/2017] [Indexed: 01/02/2023] Open
Abstract
Trastuzumab-induced cardiotoxicity (TIC) is the primary adverse event that limits the use of trastuzumab in HER2-positive breast cancer patients. However, the incidence and risk factors of TIC in HER2-positive gastric cancer are not known. Therefore, we evaluated the incidence and predictive factors of TIC in gastric cancer patients treated with trastuzumab in clinical practice. We reviewed cardiac dysfunction in HER2-positive gastric cancer patients between December 2005 and April 2015 in a prospectively-collected database that included prospective clinical trials at Yonsei Cancer Center, Republic of Korea. TIC was defined as an absolute decline in left ventricular ejection fraction (LVEF) of at least 10 percentage points from the baseline to a value less than 55%, as identified by a multiple-gated acquisition scan or an echocardiogram. Among the 115 patients, 70 patients (60.9%) received trastuzumab combined with chemotherapy, and 45 patients (39.1%) received chemotherapy alone as a first-line therapy. Symptomatic heart failure was not observed in either group, but a significant asymptomatic drop in LVEF was noted in five (7.1%) of the trastuzumab combined-group patients and in one (2.2%) chemotherapy-only group patient [hazard ratio (HR), 3.47; 95% confidence interval (CI), 0.40-29.8; P=0.257]. TIC was observed more frequently in elderly patients than in younger patients (HR, per age in year, 1.16; 95% CI, 1.02-1.31; P=0.019). Similar to prior observations in breast cancer, TIC in gastric cancer patients is not frequent or reversible. However, the asymptomatic drop in LVEF should be monitored continually in HER2-positive gastric cancer patients treated with trastuzumab, especially in elderly patients.
Collapse
|
68
|
Walsh KS, Paltin I, Gioia GA, Isquith P, Kadan-Lottick NS, Neglia JP, Brouwers P. Everyday executive function in standard-risk acute lymphoblastic leukemia survivors. Child Neuropsychol 2014; 21:78-89. [PMID: 24428397 DOI: 10.1080/09297049.2013.876491] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We aimed to evaluate parent-rated executive function (EF) in pediatric standard risk acute lymphoblastic leukemia (SR-ALL) survivors compared to a healthy comparison (HC) group. We hypothesized that SR-ALL survivors would have greater reported executive dysfunction compared to HC, and that those younger at the time of treatment would demonstrate greater EF difficulties. A sample of 256 SR-ALL survivors evaluated an average nine years after treatment were compared to HC matched for gender, assessment age, and maternal education. Profile analysis was used to compare the groups across EF scales on the BRIEF. The prevalence of clinical elevations in the groups was compared via chi square, and odds ratios were calculated. Regression models were applied to examine the role of age at diagnosis and age at assessment in reported EF. Results indicated that SR-ALL survivors' mean scores of EF are similar to HC, except for flexibility and initiation. Survivors were rated as having clinical impairments with flexibility, initiation, working memory, and emotional control at rates two to three times that of HC. The risk of working memory and self-monitoring deficits was greater in survivors who were older when assessed. There was no relationship between age at diagnosis or treatment regimen on EF. These findings suggest sparing of extensive and severe EF deficits in SR-ALL survivors overall. However, a subset of survivors displays clinically significant executive dysfunction. There appears to be a heightened susceptibility to disrupted metacognitive functions as survivors age. This has implications for how we monitor neurocognitive development and functioning of SR-ALL survivors, and highlights opportunities for cognitive interventions.
Collapse
Affiliation(s)
- Karin S Walsh
- a Children's National Medical Center & The George Washington University Medical Center , Washington , DC , USA
| | | | | | | | | | | | | |
Collapse
|