1
|
Storey S, Luo X, Ofner S, Perkins SM, Von Ah D. The role of glycemic control and symptoms and symptom clusters in breast cancer survivors with type 2 diabetes. Support Care Cancer 2025; 33:371. [PMID: 40210821 PMCID: PMC11985596 DOI: 10.1007/s00520-025-09434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
PURPOSE The purpose of the study was to describe the type and number of symptoms and examine symptom clusters of breast cancer survivors (BCS) with diabetes (type 2) by glycemic control (HbA1c < 7 or ≥ 7%). METHODS A retrospective cohort study was conducted. Symptom data were extracted from clinical notes in the electronic health record. BCS (stage I-III) diagnosed between 2007 and 2019 had diabetes, and at least one HbA1c within 8 months of initial chemotherapy was included. Zero-inflated negative binomial regression analysis was used to examine total symptoms by glycemic control. Exploratory factor analysis was conducted to identify symptom clusters. RESULTS Three hundred twenty-seven BCS met the inclusion criteria. Two symptom clusters were identified in BCS with HbA1c ≥ 7%: a psychoneurological cluster (anxiety, fatigue, peripheral neuropathy, and depression) and a gastrointestinal cluster (vomiting, nausea, and constipation). Two symptom clusters were identified in BCS with HbA1c < 7% a mixed gastrointestinal/psychoneurological cluster (vomiting, nausea, peripheral neuropathy, fatigue, and constipation) and a mental health symptom cluster (depression and anxiety). CONCLUSION The symptom clusters of BCS differed by glycemic control. Prospective research studies are needed to examine the role of glycemic control in symptoms in BCS with diabetes. Understanding the influence of glycemic control can help providers identify BCS at high risk for troublesome symptoms and symptom clusters, thereby facilitating interventions that target glycemic control, potentially mitigating symptoms, and symptom clusters, and improving outcomes for BCS with diabetes.
Collapse
Affiliation(s)
- Susan Storey
- Indiana University School of Nursing, Indianapolis, IN, 46260, USA.
| | - Xiao Luo
- Department of Management Science and Information Systems, School of Business, Oklahoma State University, Stillwater, OK, USA
| | - Susan Ofner
- Department of Biostatistics and Health Data Science, School of Medicine and Richard M. Fairbanks School of Public Health, Indiana University, 410 W 10 th Street, Suite 3000, Indianapolis, IN, 46202, USA
| | - Susan M Perkins
- Department of Biostatistics and Health Data Science, School of Medicine and Richard M. Fairbanks School of Public Health, Indiana University, 410 W 10 th Street, Suite 3000, Indianapolis, IN, 46202, USA
| | - Diane Von Ah
- Cancer Research, Center for Healthy Aging, Self-Management and Complex Care, College of Nursing, The Ohio State University (OSU), 394 Newton Hall, 1585 Neil Avenue, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, Cancer Control Program, OSU, 394 Newton Hall, 1585 Neil Avenue, Columbus, OH, 43210, USA
| |
Collapse
|
2
|
Kim J, Lim KH. Prognostic Factors for Hyperglycemia in Patients Receiving Chemotherapy. Cancer Nurs 2025; 48:112-120. [PMID: 37991472 DOI: 10.1097/ncc.0000000000001271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
BACKGROUND Approximately 10% to 30% of patients who receive chemotherapy experience hyperglycemia, which can affect the adverse reactions and treatment efficacy of chemotherapy. However, there is a paucity of research to explore the factors affecting hyperglycemia and include them in nursing interventions. OBJECTIVE The aim of this study was to understand the prognostic factors of hyperglycemia in cancer patients on chemotherapy. METHODS This retrospective, descriptive study included 134 adult patients with cancer receiving chemotherapy at Keimyeng University Dongsan Hospital in Daegu between July 1, 2021, and March 31, 2022. Data were analyzed using frequency, percentage, average, and standard deviation statistics and compared by t test, χ 2 test, and logistic regression analysis. RESULTS Logistic regression analysis revealed that sex (male) (95% confidence interval [CI], 7.24-745.49; odds ratio [OR], 73.48); education (95% CI, 4.02-201.59; OR, 28.46); exercise durations of 30 to 60 minutes (95% CI, 0.00-0.06; OR, 0.01), 60 to 90 minutes (95% CI, 0.00-0.05, OR = 0.01), and ≥90 minutes (95% CI, 0.00-0.42; OR, 0.03); exercise amount ≥ 2000 kcal/wk (95% CI, 0.01-0.77; OR, 0.06); and daily fat (95% CI, 1.02-1.14; OR, 1.08), protein (95% CI, 0.82-0.95; OR, 0.88), and seaweed (95% CI, 0.85-0.99; OR, 0.92) intake were significant predictors of hyperglycemia. CONCLUSION The prognostic factors of hyperglycemia should be included in nursing interventions to prevent and manage hyperglycemia, which in turn may help reduce adverse reactions related to chemotherapy and improve treatment efficacy. IMPLICATIONS FOR PRACTICE To prevent and manage hyperglycemia in patients on chemotherapy, prognostic factors, including exercise and protein, fat, and seaweed consumption, should be considered in nursing interventions. Particularly, in men and patients with low education levels who are at a high risk of hyperglycemia, nursing interventions for diet and exercise should be individualized.
Collapse
Affiliation(s)
- Jiyeong Kim
- Author Affiliations: College of Nursing (Ms Kim) and Department of Nursing (Dr Lim), Keimyung University, Daegu, South Korea
| | | |
Collapse
|
3
|
Prasad K, Hegde S, Rao S, D'souza RK, George T, Suresh S, Baliga MS. Usefulness of Indian Diabetes Risk Score in Predicting Treatment-Induced Hyperglycemia in Women Undergoing Adjuvant Chemotherapy for Breast Cancer. South Asian J Cancer 2025; 14:4-14. [PMID: 40124160 PMCID: PMC11925627 DOI: 10.1055/s-0043-1775805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
In the curative treatment of cancer with adjuvant chemotherapy, antineoplastic drugs, along with glucocorticoids, can induce hyperglycemia. The objective of this study was to assess the utility of the Indian Diabetes Risk Score (IDRS) in predicting treatment-induced hyperglycemia in women who were nondiabetic and normoglycemic at the start of chemotherapy. This prospective study was conducted with nondiabetic women who required adjuvant chemotherapy. Participants voluntarily completed the IDRS, providing information on age, waist circumference, family history of diabetes, and physical activity. Chemotherapy-induced hyperglycemia was defined as fasting blood glucose levels ≥100 mg/dL or random blood glucose levels ≥140 mg/dL during treatment. Data were categorized into women who developed hyperglycemia and those who remained normoglycemic during treatment and were analyzed using Fisher's exact test. A significance level of p < 0.05 was applied. Receiver operating characteristic (ROC) curves were constructed to validate the IDRS for predicting hyperglycemia. A total of 208 women met the inclusion criteria and participated in the study. The results revealed that 38.93% (81/208) developed hyperglycemia by the end of chemotherapy, as observed during their first follow-up after treatment. Fisher's exact test demonstrated a significant difference in the total IDRS score and its domains, including family history, physical activity, and waist circumference ( p = 0.017-< 0.001), but not age. ROC analysis indicated that an IDRS score above 60 increased the likelihood of developing hyperglycemia, with a sensitivity of 81.3%, specificity of 54.7%, and an area under the curve of 0.727. These findings suggest that the IDRS is a sensitive tool for predicting adjuvant chemotherapy-induced hyperglycemia in breast cancer patients without diabetes. To the best of the authors' knowledge, this is the first study to evaluate the utility of the IDRS in predicting treatment-induced hyperglycemia in women undergoing adjuvant chemotherapy for breast cancer. Ongoing efforts are focused on understanding the underlying mechanisms and strategies for mitigation.
Collapse
Affiliation(s)
- Krishna Prasad
- Department of Medical Oncology, Mangalore Institute of Oncology, Mangaluru, Karnataka, India
| | - Sanath Hegde
- Department of Radiation Oncology, Mangalore Institute of Oncology, Mangaluru, Karnataka, India
| | - Suresh Rao
- Department of Radiation Oncology, Mangalore Institute of Oncology, Mangaluru, Karnataka, India
| | - Rhea Katherine D'souza
- Department of Research, Research Unit, Mangalore Institute of Oncology, Mangaluru, Karnataka, India
| | - Thomas George
- Department of Research, Research Unit, Mangalore Institute of Oncology, Mangaluru, Karnataka, India
| | - Sucharitha Suresh
- Department of Community Medicine, Father Muller Medical College, Mangalore, Karnataka, India
| | | |
Collapse
|
4
|
Taguchi A, Kato K, Furusawa A, Hara K, Sone K, Yamada K, Kajiyama H, Shimada M, Okamoto A. Heterogeneous treatment effect of dose-dense paclitaxel plus carboplatin therapy for advanced ovarian cancer. Int J Cancer 2024; 155:1068-1077. [PMID: 38712630 DOI: 10.1002/ijc.34996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
A Japanese clinical trial (JGOG3016) showed that dose-dense weekly paclitaxel in combination with carboplatin extensively prolonged overall survival (OS) in patients with advanced ovarian cancer. However, in other clinical trials, dose-dense paclitaxel regimens were not superior to triweekly paclitaxel regimens. In this study, causal tree analysis was applied to explore subpopulations with different treatment effects of dose-dense paclitaxel in a data-driven approach. The 587 participants with stage II-IV ovarian cancer in the JGOG3016 trial were used for model development. The primary endpoint was treatment effect in terms of 3-year OS in patients receiving dose-dense vs. conventional paclitaxel therapies. In patients <50 years, the 3-year OS was similar in both groups; however, it was higher in the dose-dense group in patients ≥50 years. Dose-dense paclitaxel showed strong positive treatment effects in patients ≥50 years with stage II/III disease, BMI <23 kg/m2, non-CC/MC, and residual tumor ≥1 cm. In contrast, although there was no significant difference in OS; the 3-year OS rate was 23% lower in dose-dense paclitaxel than conventional paclitaxel in patients ≥60 years with stage IV cancer. Patients in this group had a particularly lower performance status than other groups. Our causal tree analysis suggested that poor prognosis groups represented by residual tumor tissue ≥1 cm benefit from dose-dense paclitaxel, whereas elderly patients with advanced disease and low-performance status are negatively impacted by dose-dense paclitaxel. These subpopulations will be of interest to future validation studies. Personalized treatments based on clinical features are expected to improve advanced ovarian cancer prognosis.
Collapse
Affiliation(s)
- Ayumi Taguchi
- Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Bunkyo-ku, Tokyo, Japan
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center Osaka University, Suita-shi, Osaka, Japan
| | - Kosuke Kato
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Akiko Furusawa
- Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Bunkyo-ku, Tokyo, Japan
- Department of Gynecology, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka, Japan
| | - Konan Hara
- Department of Economics, University of Arizona, Tucson, Arizona, USA
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kyosuke Yamada
- Department of Obstetrics and Gynecology, Daisan Hospital, The Jikei University School of Medicine, Komae, Tokyo, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Muneaki Shimada
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi, Japan
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Salgado TM, Birari PB, Alshahawey M, Hickey Zacholski E, Mackler E, Buffington TM, Musselman KT, Irvin WJ, Perkins JM, Le TN, Dixon DL, Farris KB, Sheppard VB, Jones RM. Optimal hyperglycemia thresholds in patients undergoing chemotherapy: a cross sectional study of oncologists' practices. Support Care Cancer 2024; 32:563. [PMID: 39088060 PMCID: PMC11294379 DOI: 10.1007/s00520-024-08756-0] [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: 01/14/2024] [Accepted: 07/21/2024] [Indexed: 08/02/2024]
Abstract
PURPOSE Neither the United States nor the European oncology guidelines include details for appropriate management of hyperglycemia in cancer patients. The aim was to identify fasting and random blood glucose thresholds, and hemoglobin A1c (HbA1c) targets used by oncologists in clinical practice when managing hyperglycemia in patients with cancer undergoing chemotherapy. METHODS This national, cross sectional study utilized a questionnaire to collect oncologists' perceptions about optimal blood glucose thresholds and HbA1c targets in patients with cancer undergoing chemotherapy. Descriptive statistics were calculated to summarize glucose thresholds, HbA1c targets, and sample characteristics. Responses to an open-ended question about oncologists' approach to hyperglycemia management were analyzed via thematic analysis using an inductive approach. RESULTS Respondents (n = 229) were on average 52.1 years of age, 67.7% men, and 91.3% White. For patients without diabetes but experiencing hyperglycemia, oncologists targeted lower and upper fasting blood glucose levels between 75-121 mg/dL and 105-135 mg/dL, respectively. For patients with diabetes, the targets for lower and upper fasting blood glucose levels ranged between 100-130 mg/dL and 128-150 mg/dL, respectively. Fasting blood glucose (95.6%) and HbA1c (78.6%) were the most commonly used clinical indicators to consider chemotherapy dose reduction, delay, or discontinuation due to hyperglycemia in patients receiving chemotherapy with curative intent. Among those receiving palliative intent chemotherapy, the preferred clinical parameters were random blood glucose (90.0%), patient-reported blood glucose readings (70.7%), continuous glucose monitoring readings (65.1%), and patient-reported symptoms of hyperglycemia (65.1%). Three main themes emerged about oncologists' approach to hyperglycemia management: 1) identification of high-risk patients; 2) need for early identification, screening, and diagnosis of hyperglycemia; and 3) multiple hyperglycemia management strategies. CONCLUSION Oncologists reported a wide variation of target blood glucose ranges considered appropriate in patients undergoing chemotherapy. Lack of clear guidance for hyperglycemia management during chemotherapy in the United States may be contributing to a lack of consistency in clinical practice.
Collapse
Affiliation(s)
- Teresa M Salgado
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy and Massey Cancer Center, Virginia Commonwealth University, 410 N. 12Th Street PO Box 98053, Richmond, VA, 23298, USA.
| | - Poorva B Birari
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, 410 N. 12Th Street PO Box 98053, Richmond, VA, 23298, USA
| | - Mona Alshahawey
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, 410 N. 12Th Street PO Box 98053, Richmond, VA, 23298, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St, El-Qobba Bridge, El Weili, Cairo Governorate, 4393005, Egypt
| | - Erin Hickey Zacholski
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, 410 N. 12Th Street PO Box 98053, Richmond, VA, 23298, USA
| | - Emily Mackler
- Michigan Oncology Quality Consortium (MOQC) and Michigan Institute for Care Management and Transformation (MICMT), 4251 Plymouth Road Arbor Lakes, Building 3, Floor 3, Ann Arbor, MI, 48105, USA
| | - Tonya M Buffington
- Bon Secours Mercy Health, 611 Watkins Centre Parkway, Suite 250, Midlothian, VA, 23114, USA
| | - Kerri T Musselman
- Emcara Health and PopHealthCare, 113 Seaboard Lane, Suite B200, Franklin, TN, 37067, USA
| | - William J Irvin
- Bon Secours St Francis, 14051 St Francis Blvd Suite 2210, Midlothian, VA, 23114, USA
| | - Jennifer M Perkins
- Division of Endocrinology, University of California San Francisco Medical Center, 400 Parnassus Ave., Suite A-550, San Francisco, CA, 94143, USA
| | - Trang N Le
- Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, 1101 E. Marshall St. Sanger Hall Suite 1-030, Richmond, VA, 23298, USA
| | - Dave L Dixon
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, 410 N. 12Th Street PO Box 98053, Richmond, VA, 23298, USA
| | - Karen B Farris
- Department of Clinical Pharmacy Translational Sciences, College of Pharmacy and Michigan Institute for Care Management and Transformation (MICMT), University of Michigan, 428 Church St, Ann Arbor, MI, 48109, USA
| | - Vanessa B Sheppard
- Department of Social and Behavioral Sciences, School of Public Health, and Massey Cancer Center, Virginia Commonwealth University, 830 East Main Street, Richmond, VA, 23219, USA
| | - Resa M Jones
- Department of Epidemiology and Biostatistics, College of Public Health, and Fox Chase Cancer Center, Temple University, 1301 Cecil B. Moore Avenue Ritter Annex, 9Th Floor, Suite 917, Philadelphia, PA, 19122, USA
| |
Collapse
|
6
|
Ziegengeist JL, Elmes JB, Strassels SA, Patel JN, Moore DC. Alpelisib-Induced Diabetic Ketoacidosis: A Pharmacovigilance Analysis of the FDA Adverse Event Reporting System and Review of the Literature. Clin Breast Cancer 2024; 24:e204-e209. [PMID: 38245400 DOI: 10.1016/j.clbc.2024.01.004] [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: 10/24/2023] [Revised: 12/26/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Alpelisib is a PI3K inhibitor indicated with fulvestrant for treatment of advanced or metastatic hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative, PIK3CA-mutated breast cancer. In the phase III SOLAR-1 trial, grade 3/4 hyperglycemic events were reported in 36.6% of patients receiving alpelisib-fulvestrant compared to 0.7% receiving placebo-fulvestrant. As case reports of diabetic ketoacidosis (DKA) have been associated with alpelisib use, the goal of this study was to characterize the FAERS reported cases of this severe adverse effect. METHODS A retrospective disproportionality analysis was performed using the FAERS database by calculating the reporting odds ratio (ROR) of DKA events with alpelisib from 2019 to 2022. A PubMed literature review of case reports characterizing alpelisib-induced DKA was performed. RESULTS Pharmacovigilance database analysis revealed significance in reporting among 87 DKA cases with alpelisib (ROR 9.84, 95% confidence interval 7.3-13.2), including hospitalization and death as reported outcomes. Review of 11 published case reports reveals median onset of DKA at 14 days with successful rechallenge possible. CONCLUSION Significant association with reporting exists between DKA and alpelisib exposure. We observed similar median time to onset of hyperglycemia between our analysis compared to that reported in SOLAR-1. Considering early onset of this toxicity, it is imperative that patients be closely monitored when initiating alpelisib. Addition of a preemptive antihyperglycemic or escalation in those previously on antihyperglycemic medications is beneficial in decreasing the severity of hyperglycemia with alpelisib. Further study investigating risk factors is warranted to better elucidate which patients require preemptive therapy.
Collapse
Affiliation(s)
- Julia L Ziegengeist
- Department of Pharmacy, Clinical Pharmacist Coordinator, Levine Cancer Institute, Atrium Health, Charlotte, NC.
| | - Joseph B Elmes
- Department of Pharmacy, Oncology Clinical Staff Pharmacist, Levine Cancer Institute, Atrium Health, Concord, NC
| | | | - Jai N Patel
- Department of Cancer Pharmacology and Pharmacogenomics, Clinical Pharmacology and Pharmacogenomics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Donald C Moore
- Department of Pharmacy, Clinical Oncology Pharmacy Manager, Levine Cancer Institute, Atrium Health, Charlotte, NC
| |
Collapse
|
7
|
Harrod JC, Cheung YMM, Buckley L, Cromwell GE, Fowler KM, Hughes ME, Lin NU, Tolaney SM, Min L, McDonnell ME. Impact of personalized diabetes care on distress and treatment satisfaction in people with breast cancer. Diabet Med 2024; 41:e15292. [PMID: 38291604 DOI: 10.1111/dme.15292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
AIMS In patients with breast cancer (BCa) and diabetes (DM), diabetes distress (DD) and treatment satisfaction (DTS) can influence BCa management and outcomes. We assessed the impact of implementing a personalized diabetes care model in patients with BCa. METHODS Patients in active treatment or surveillance for BCa with an HbA1c > 53 mmol/mol (7%) or random blood glucose >11.1 mmol/L were included. Participants were offered continuous glucose monitoring (CGM), virtual care and a dedicated diabetes provider for 6 months. Primary outcomes included DD measured by the Diabetes Distress Survey (DDS) and DTS measured by the Diabetes Treatment Satisfaction Questionnaire (DTSQ). Questionnaires were conducted at 0, 3 and 6 months. RESULTS Thirty-one women were enrolled (median age 61, IQR 49.0-69.0). Compared to baseline, the mean DDS score was lower at both 3 months (2.2 vs. 1.8 [n = 27], p = 0.004, SD = 0.70) and 6 months (2.3 vs. 1.8 [n = 23], p = 0.002, SD = 0.70). The mean DTSQ score was higher at 3 months (baseline: 20.5 vs. 3 months: 28.7 [n = 28], p < 0.001, SD = 9.2) and 6 months (baseline: 20.4 vs. 6 months: 30.0 [n = 26], p < 0.001, SD = 9.7). CONCLUSIONS Personalized diabetes care models that emphasize remote management and optimize access for those with BCa may lower DD and improve DTS.
Collapse
Affiliation(s)
- Julia C Harrod
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Yee-Ming M Cheung
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Endocrinology, Diabetes and Metabolism, Northwell Health, Manhasset, New York, USA
| | - Lauren Buckley
- Division of Breast Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Grace E Cromwell
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kristen M Fowler
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Melissa E Hughes
- Division of Breast Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nancy U Lin
- Division of Breast Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sara M Tolaney
- Division of Breast Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Le Min
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Marie E McDonnell
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Salgado TM, Radwan RM, Hickey Zacholski E, Mackler E, Buffington TM, Musselman KT, Irvin WJ, Perkins JM, Le TN, Dixon DL, Farris KB, Sheppard VB, Jones RM. Oncologists' responsibility, comfort, and knowledge managing hyperglycemia in patients with cancer undergoing chemotherapy: a cross sectional study. Support Care Cancer 2023; 31:450. [PMID: 37421495 DOI: 10.1007/s00520-023-07927-9] [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: 03/16/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023]
Abstract
PURPOSE To assess oncologists' responsibility, comfort, and knowledge managing hyperglycemia in patients undergoing chemotherapy. METHODS In this cross-sectional study, a questionnaire collected oncologists' perceptions about professionals responsible for managing hyperglycemia during chemotherapy; comfort (score range 12-120); and knowledge (score range 0-16). Descriptive statistics were calculated including Student t-tests and one-way ANOVA for mean score differences. Multivariable linear regression identified predictors of comfort and knowledge scores. RESULTS Respondents (N = 229) were 67.7% men, 91.3% White and mean age 52.1 years. Oncologists perceived endocrinologists/diabetologists and primary care physicians as those responsible for managing hyperglycemia during chemotherapy, and most frequently referred to these clinicians. Reasons for referral included lack of time to manage hyperglycemia (62.4%), belief that patients would benefit from referral to an alternative provider clinician (54.1%), and not perceiving hyperglycemia management in their scope of practice (52.4%). The top-3 barriers to patient referral were long wait times for primary care (69.9%) and endocrinology (68.1%) visits, and patient's provider outside of the oncologist's institution (52.8%). The top-3 barriers to treating hyperglycemia were lack of knowledge about when to start insulin, how to adjust insulin, and what insulin type works best. Women (ß = 1.67, 95% CI: 0.16, 3.18) and oncologists in suburban areas (ß = 6.98, 95% CI: 2.53, 11.44) had higher comfort scores than their respective counterparts; oncologists working in practices with > 10 oncologists had lower comfort scores (ß = -2.75, 95% CI: -4.96, -0.53) than those in practices with ≤ 10. No significant predictors were identified for knowledge. CONCLUSION Oncologists expected endocrinology or primary care clinicians to manage hyperglycemia during chemotherapy, but long wait times were among the top barriers cited when referring patients. New models that provide prompt and coordinated care are needed.
Collapse
Affiliation(s)
- Teresa M Salgado
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, and Massey Cancer Center, Virginia Commonwealth University, PO Box 98053, 410 N. 12th Street, Richmond, VA, 23298, USA.
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, PO Box 980533, 410 N. 12th Street, Richmond, VA, 23298, USA.
| | - Rotana M Radwan
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, PO Box 980533, 410 N. 12th Street, Richmond, VA, 23298, USA
| | - Erin Hickey Zacholski
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, PO Box 980533, 410 N. 12th Street, Richmond, VA, 23298, USA
| | - Emily Mackler
- Michigan Oncology Quality Consortium (MOQC) and Michigan Institute for Care Management and Transformation (MICMT), 4251 Plymouth Road Arbor Lakes, Building 3, Floor 3, Ann Arbor, MI, 48105, USA
| | - Tonya M Buffington
- Bon Secours Mercy Health, 611 Watkins Centre Parkway, Suite 250, Midlothian, Richmond, VA, 23114, USA
| | - Kerri T Musselman
- Emcara Health and PopHealthCare, 113 Seaboard Lane, Suite B200, Franklin, TN, 37067, USA
| | - William J Irvin
- Bon Secours Cancer Institute, Bon Secours Mercy Health, 14051 St Francis Blvd Suite 2210, Midlothian, VA, 23114, United States
| | - Jennifer M Perkins
- Division of Endocrinology, University of California San Francisco Medical Center, Endocrinology Clinic at Parnassus 400 Parnassus Ave., Suite A-550, San Francisco, CA, 94143, USA
| | - Trang N Le
- Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, 1101 E. Marshall St. Sanger Hall Suite 1-030, Richmond, VA, 23298, USA
| | - Dave L Dixon
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, PO Box 980533, 410 N. 12th Street, Richmond, VA, 23298, USA
| | - Karen B Farris
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI, 48109, USA
| | - Vanessa B Sheppard
- Department of Health Behavior and Policy, School of Population Health, and Massey Comprehensive Cancer Center, Virginia Commonwealth University, 830 East Main Street, Richmond, VA, 23219, USA
| | - Resa M Jones
- Department of Epidemiology & Biostatistics, College of Public Health, and Fox Chase Cancer Center, Temple University, 1301 Cecil B. Moore Avenue Ritter Annex, 9th Floor, Suite 917, Philadelphia, PA, 19122, USA
| |
Collapse
|
9
|
Hyperglycemia and Glycemic Variability Associated with Glucocorticoids in Women without Pre-Existing Diabetes Undergoing Neoadjuvant or Adjuvant Taxane Chemotherapy for Early-Stage Breast Cancer. J Clin Med 2023; 12:jcm12051906. [PMID: 36902693 PMCID: PMC10004215 DOI: 10.3390/jcm12051906] [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: 02/09/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Glucocorticoids, which are administered with chemotherapy, cause hyperglycemia. Glycemic variability among breast cancer patients without diabetes is not well known. A retrospective cohort study was conducted involving early-stage breast cancer patients without diabetes who received dexamethasone prior to neoadjuvant or adjuvant taxane chemotherapy between August 2017-December 2019. Random blood glucose levels were analyzed, and steroid-induced hyperglycemia (SIH) was defined as a random glucose level of >140 mg/dL. A multivariate proportional hazards model was used to identify the risk factors of SIH. Out of 100 patients, the median age was 53 years (IQR: 45-63.5). A total of 45% of patients were non-Hispanic White, 28% Hispanic, 19% Asian, and 5% African American. The incidence of SIH was 67%, and glycemic fluctuations were highest in those with glucose levels of >200 mg/dL. Non-Hispanic White patients represented a significant predictor for time to SIH, with a hazard ratio of 2.5 (95% CI: 1.04, 5.95, p = 0.039). SIH was transient in over 90% of the patients, and only seven patients remained hyperglycemic after glucocorticoid and chemotherapy completion. Pretaxane dexamethasone-induced hyperglycemia was observed in 67% of the patients, with the greatest glycemic lability in those patients with blood glucose levels of >200 mg/dL. The non-Hispanic White patients had a higher risk of developing SIH.
Collapse
|
10
|
Chan DS, Vieira R, Abar L, Aune D, Balducci K, Cariolou M, Greenwood DC, Markozannes G, Nanu N, Becerra‐Tomás N, Giovannucci EL, Gunter MJ, Jackson AA, Kampman E, Lund V, Allen K, Brockton NT, Croker H, Katsikioti D, McGinley‐Gieser D, Mitrou P, Wiseman M, Cross AJ, Riboli E, Clinton SK, McTiernan A, Norat T, Tsilidis KK. Postdiagnosis body fatness, weight change and breast cancer prognosis: Global Cancer Update Program (CUP global) systematic literature review and meta-analysis. Int J Cancer 2023; 152:572-599. [PMID: 36279884 PMCID: PMC10092239 DOI: 10.1002/ijc.34322] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 07/29/2022] [Accepted: 09/05/2022] [Indexed: 02/01/2023]
Abstract
Previous evidence on postdiagnosis body fatness and mortality after breast cancer was graded as limited-suggestive. To evaluate the evidence on body mass index (BMI), waist circumference, waist-hip-ratio and weight change in relation to breast cancer prognosis, an updated systematic review was conducted. PubMed and Embase were searched for relevant studies published up to 31 October, 2021. Random-effects meta-analyses were conducted to estimate summary relative risks (RRs). The evidence was judged by an independent Expert Panel using pre-defined grading criteria. One randomized controlled trial and 225 observational studies were reviewed (220 publications). There was strong evidence (likelihood of causality: probable) that higher postdiagnosis BMI was associated with increased all-cause mortality (64 studies, 32 507 deaths), breast cancer-specific mortality (39 studies, 14 106 deaths) and second primary breast cancer (11 studies, 5248 events). The respective summary RRs and 95% confidence intervals per 5 kg/m2 BMI were 1.07 (1.05-1.10), 1.10 (1.06-1.14) and 1.14 (1.04-1.26), with high between-study heterogeneity (I2 = 56%, 60%, 66%), but generally consistent positive associations. Positive associations were also observed for waist circumference, waist-hip-ratio and all-cause and breast cancer-specific mortality. There was limited-suggestive evidence that postdiagnosis BMI was associated with higher risk of recurrence, nonbreast cancer deaths and cardiovascular deaths. The evidence for postdiagnosis (unexplained) weight or BMI change and all outcomes was graded as limited-no conclusion. The RCT showed potential beneficial effect of intentional weight loss on disease-free-survival, but more intervention trials and well-designed observational studies in diverse populations are needed to elucidate the impact of body composition and their changes on breast cancer outcomes.
Collapse
Affiliation(s)
- Doris S.M. Chan
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Rita Vieira
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Leila Abar
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Dagfinn Aune
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- Department of NutritionBjørknes University CollegeOsloNorway
- Department of Endocrinology, Morbid Obesity and Preventive MedicineOslo University HospitalOsloNorway
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska InstitutetStockholmSweden
| | - Katia Balducci
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Margarita Cariolou
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Darren C. Greenwood
- Leeds Institute for Data Analytics, Faculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - Georgios Markozannes
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- Department of Hygiene and EpidemiologyUniversity of Ioannina Medical SchoolIoanninaGreece
| | - Neesha Nanu
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Nerea Becerra‐Tomás
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Edward L. Giovannucci
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Department of Nutrition, Harvard T. H. Chan School of Public HealthBostonMassachusettsUSA
| | - Marc J. Gunter
- Nutrition and Metabolism Section, International Agency for Research on CancerLyonFrance
| | - Alan A. Jackson
- Faculty of Medicine, School of Human Development and HealthUniversity of SouthamptonSouthamptonUK
- National Institute of Health Research Cancer and Nutrition CollaborationSouthamptonUK
| | - Ellen Kampman
- Division of Human Nutrition and HealthWageningen University & ResearchWageningenThe Netherlands
| | - Vivien Lund
- World Cancer Research Fund InternationalLondonUK
| | - Kate Allen
- World Cancer Research Fund InternationalLondonUK
| | | | - Helen Croker
- World Cancer Research Fund InternationalLondonUK
| | | | | | | | | | - Amanda J. Cross
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Elio Riboli
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Steven K. Clinton
- Division of Medical Oncology, The Department of Internal MedicineCollege of Medicine and Ohio State University Comprehensive Cancer Center, Ohio State UniversityColumbusOhioUSA
| | - Anne McTiernan
- Division of Public Health SciencesFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Teresa Norat
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- World Cancer Research Fund InternationalLondonUK
| | - Konstantinos K. Tsilidis
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- Department of Hygiene and EpidemiologyUniversity of Ioannina Medical SchoolIoanninaGreece
| |
Collapse
|
11
|
Impact of diabetes (type 2) and glycemic control on health-related outcomes of patients receiving chemotherapy for non-metastatic breast cancer: a retrospective analysis. Support Care Cancer 2023; 31:114. [PMID: 36637522 DOI: 10.1007/s00520-022-07563-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE To examine the impact of diabetes (type 2) and glycemic control on healthcare-related outcomes (healthcare utilization, adverse effects, and treatment modifications) in non-metastatic breast cancer (NMBC) patients during chemotherapy treatment. METHODS This was a retrospective study of 243 NMBC patients (stages 1-3) with/without diabetes receiving neoadjuvant or adjuvant cytotoxic chemotherapy. The primary study endpoint was to compare healthcare utilization between NMBC patients with and without diabetes. Secondary study endpoints included adverse events and chemotherapy treatment modifications. Additional analyses were conducted to compare these health-related outcomes by glycemic control status. RESULTS NMBC patients with diabetes had higher utilization of emergency department (ED) services (52% vs. 33%, p = 0.013) and a higher frequency of unplanned inpatient admissions (35% vs. 19%, p = 0.014). Additionally, NMBC patients with diabetes had a higher incidence of infection and treatment modifications. NMBC patients, regardless of diabetes diagnosis, who had poor glycemic control, specifically hyperglycemia (per random blood glucose), during the study period also had increased healthcare utilization, adverse effects, and treatment modifications. Patients with a baseline HbA1c ≥ 7 had a greater number of ED visits and a higher incidence of infection than those without diabetes. CONCLUSION Diabetes and glycemic control may impact the health-related outcomes of NMBC patients. Additional studies are needed to confirm these findings and determine optimal monitoring and management strategies for NMBC patients with diabetes and/or poor glycemic control during cytotoxic chemotherapy.
Collapse
|
12
|
Zimbalist AS, Caan BJ, Chen WY, Mittendorf EA, Dillon DAR, Quesenberry C, Cespedes Feliciano EM. Metabolic abnormalities and survival among patients with non-metastatic breast cancer. BMC Cancer 2022; 22:1361. [PMID: 36581817 PMCID: PMC9801571 DOI: 10.1186/s12885-022-10430-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/09/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Research on the impact of metabolic abnormalities on breast cancer prognosis is limited by small samples and assessment of laboratory values at a single time point, often prior to cancer diagnosis and treatment. In this population-based cohort, time-updated laboratory values were adjusted for cancer treatment to assess the association between metabolic risk factors (glucose, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglycerides) and breast cancer survival. METHODS 13,434 women diagnosed with stage I-III breast cancer from 2005-15 at Kaiser Permanente were included. All outpatient fasting glucose, HDL-C, LDL-C, and triglyceride values from diagnosis through 2019 or death were extracted from electronic medical records. Risk of breast cancer-specific mortality was evaluated with Cox proportional hazards models adjusted for metabolic labs, demographics, body mass index, diabetes, dyslipidemia and anti-hypertensive medications, tumor characteristics (stage, ER and HER2 receptor status) and cancer treatment (use of chemotherapy, tamoxifen, and aromatase inhibitors). RESULTS Mean (SD) age at diagnosis was 62.3 (11.8) years. Over a median follow-up of 8.6 years, 2,876 patients died; 1,080 of breast cancer. Patients with low HDL-C (≤ 45 vs. > 45 mg/dL) had higher breast cancer-specific mortality (HR, 1.77; 95% CI, 1.53-2.05), as did those with elevated fasting glucose (> 99 vs. 60-99 mg/dL) (HR, 1.19; 95% CI, 1.03-1.37). Elevated levels of triglycerides and LDL-C were not associated with breast cancer-specific mortality. CONCLUSIONS High fasting glucose and low HDL-C evaluated over time after cancer diagnosis were associated with higher breast cancer mortality independent of cancer treatments and changes in other metabolic risk factors. Future studies should address whether pharmacologic or lifestyle treatment of glucose and lipids after breast cancer diagnosis can optimize survival outcomes.
Collapse
Affiliation(s)
- Alexa S. Zimbalist
- grid.280062.e0000 0000 9957 7758Division of Research, Kaiser Permanente Northern California, 2000 Broadway, 5Th Floor, Oakland, CA 94612 USA
| | - Bette J. Caan
- grid.280062.e0000 0000 9957 7758Division of Research, Kaiser Permanente Northern California, 2000 Broadway, 5Th Floor, Oakland, CA 94612 USA
| | - Wendy Y. Chen
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02115 USA ,grid.65499.370000 0001 2106 9910Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215 USA
| | - Elizabeth A. Mittendorf
- grid.62560.370000 0004 0378 8294Division of Breast Surgery, Brigham and Women’s Hospital, Boston, MA 02215 USA ,grid.65499.370000 0001 2106 9910Breast Oncology, Dana-Farber Brigham Cancer Center, Boston, MA 02215 USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA 02215 USA
| | - Deborah A. R. Dillon
- grid.38142.3c000000041936754XHarvard Medical School, Boston, MA 02215 USA ,grid.62560.370000 0004 0378 8294Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Charles Quesenberry
- grid.280062.e0000 0000 9957 7758Division of Research, Kaiser Permanente Northern California, 2000 Broadway, 5Th Floor, Oakland, CA 94612 USA
| | - Elizabeth M. Cespedes Feliciano
- grid.280062.e0000 0000 9957 7758Division of Research, Kaiser Permanente Northern California, 2000 Broadway, 5Th Floor, Oakland, CA 94612 USA
| |
Collapse
|
13
|
Joharatnam-Hogan N, Morganstein DL. Diabetes and Cancer - optimising glycaemic control. J Hum Nutr Diet 2022; 36:504-513. [PMID: 35748508 DOI: 10.1111/jhn.13051] [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: 03/02/2022] [Accepted: 05/31/2022] [Indexed: 01/08/2023]
Abstract
Diabetes and cancer are both common and increasingly prevalent conditions, but emerging epidemiological evidence confirms that the risk of developing a number of common cancers is increased in those with type 2 diabetes. The risk of cancer in type 1 diabetes is less clearly defined, and therefore this review will focus on type 2 diabetes. Emerging evidence also supports an influence of diabetes on outcomes of cancer treatment. However, this relationship is bi-directional, with cancer and its treatment impacting on glucose control, whilst there is also emerging evidence that diabetes care can deteriorate after a cancer diagnosis (summarised in Figure 1). Despite these clear links there is a lack of evidence to guide clinicians in how to manage patients with diabetes during their cancer treatment. Although recent UK guidelines have started to address this, with the development of guidance for the management of hyperglycaemia in cancer, there is a clear need for wider guidance on the management of multi-morbidity during cancer, including diabetes and obesity, to incorporate nutritional management We have therefore undertaken a narrative review of the evidence of links between type 2 diabetes and cancer incidence and outcomes, and discuss the challenges to diabetes care during cancer treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Daniel L Morganstein
- Royal Marsden Hospital, Fulham Roal, London, SW3 6JJ, UK.,Chelsea and Westminster Hospital, 369 Fulham Road, London, SW10 9NH, UK
| |
Collapse
|
14
|
Chen Y, Xu M, Ye Q, Xiang J, Xue T, Yang T, Liu L, Yan B. Irregular delay of adjuvant chemotherapy correlated with poor outcome in stage II-III colorectal cancer. BMC Cancer 2022; 22:670. [PMID: 35715761 PMCID: PMC9206266 DOI: 10.1186/s12885-022-09767-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/10/2022] [Indexed: 11/10/2022] Open
Abstract
AIMS Adjuvant chemotherapy (ACT) plays an important role in improving the survival of stage II-III colorectal cancer (CRC) patients after curative surgery. However, the prognostic role of irregular delay of ACT (IDacT) for these patients has been less studied. MATERIALS AND METHODS A total of 117 stage II-III CRC patients who underwent radical resection and received at least 3 months ACT were enrolled retrospectively. The significance of IDacT, including total delay (TD) and delay per cycle (DpC), in predicting disease-free survival (DFS) was determined using receiver operating characteristic curve (ROC) analysis. The survival differences between the TD, DpC-short and DpC-long subgroups were tested using Kaplan-Meier analysis, and risk factors for prognosis were determined using a Cox proportional hazards model. RESULTS Using 35.50 and 3.27 days as the optimal cut-off points for TD and DpC, respectively, ROC analysis revealed that TD and DpC had sensitivities of 43.60% and 59.00% and specificities of 83.30% and 62.80%, respectively, in predicting DFS (both P < 0.05). No differences in the clinicopathological parameters were found between the TD, DpC-short or -long subgroups except histological differentiation in different TD subgroups and combined T stages in different DpC subgroups (both P = 0.04). Patients in the TD or DpC-long group exhibited significantly worse survival than in the -short group (TD: Log rank = 9.11, P < 0.01; DpC: Log rank = 6.09, P = 0.01). DpC was an independent risk factor for prognosis (HR = 2.54, 95% CI: 1.32-4.88, P = 0.01). CONCLUSIONS IDacT had a profound effect on the outcome for stage II-III CRC. Although TD and DpC were significant for the prognosis, DpC was more robust, and patients who presented DpC for a long time had a significantly worse DFS.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of General Medicine, Hainan Hospital of Chinese PLA General Hospital, Sanya City, Hainan, P.R. China
| | - Mingyue Xu
- Department of General Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya City, Hainan, P.R. China
| | - Qianwen Ye
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya City, Hainan province, 572000, P.R. China
| | - Jia Xiang
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya City, Hainan province, 572000, P.R. China
| | - Tianhui Xue
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya City, Hainan province, 572000, P.R. China
| | - Tao Yang
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya City, Hainan province, 572000, P.R. China
| | - Long Liu
- Department Traditional Chinese Medicine, Tianyou Hospital of Tongji University, No. 528 of Zhennan Road, Putuo District, Shanghai, 200331, P.R. China.
| | - Bing Yan
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, No. 80 of Jianglin Road, Haitang District, Sanya City, Hainan province, 572000, P.R. China.
| |
Collapse
|
15
|
Factors leading to alpelisib discontinuation in patients with hormone receptor positive, human epidermal growth factor receptor-2 negative breast cancer. Breast Cancer Res Treat 2022; 192:303-311. [PMID: 35000092 DOI: 10.1007/s10549-021-06476-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/02/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE Alpelisib is a phosphoinositide-3-kinase inhibitor approved for hormone-receptor-positive, PIK3CA-mutated metastatic breast cancer. However, length of drug exposure, maximum-tolerated dose, and therefore clinical response can vary significantly outside of the trial setting. This study evaluates our center's "real world" experience with alpelisib and focuses on duration of therapy and factors associated with cancer progression. METHODS Patients receiving alpelisib at our center between 2019 and 2021 were identified. We evaluated duration of alpelisib therapy and the causative reasons for drug discontinuation. The association of drug duration and dose with subsequent cancer progression were assessed, along with the association between hyperglycemia during alpelisib therapy and cancer progression. RESULTS Sixty-two women prescribed alpelisib were included (mean age 61 years). Disease progression was the most common reason for drug discontinuation, while discontinuation within 30 days was primarily attributed to adverse events (AEs). Among those who progressed, median time to progression was longer in those on alpelisib for > 90 days compared with those on alpelisib for ≤ 90 days (187 vs. 77 days, p < 0.001). At 200 days, freedom from progression was greater for those on alpelisib for > 90 days compared to those receiving therapy for ≤ 90 days (59% vs. 19%, p = 0.001). Median blood glucose as a continuous variable was associated with disease progression (HR 1.01, 95% CI 1.00-1.02, p = 0.02). CONCLUSION While progression of disease is the largest contributor to alpelisib discontinuation, AEs are the leading cause for early drug cessation. Shorter alpelisib exposure is associated with greater cancer progression. Further studies are needed to determine the impact of sustained hyperglycemia on cancer progression.
Collapse
|
16
|
Nikmaneshi MR, Firoozabadi B, Mozafari A. Chemo-mechanistic multi-scale model of a three-dimensional tumor microenvironment to quantify the chemotherapy response of cancer. Biotechnol Bioeng 2021; 118:3871-3887. [PMID: 34133020 DOI: 10.1002/bit.27863] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/03/2023]
Abstract
Exploring efficient chemotherapy would benefit from a deeper understanding of the tumor microenvironment (TME) and its role in tumor progression. As in vivo experimental methods are unable to isolate or control individual factors of the TME, and in vitro models often cannot include all the contributing factors, some questions are best addressed with mathematical models of systems biology. In this study, we establish a multi-scale mathematical model of the TME to simulate three-dimensional tumor growth and angiogenesis and then implement the model for an array of chemotherapy approaches to elucidate the effect of TME conditions and drug scheduling on controlling tumor progression. The hyperglycemic condition as the most common disorder for cancer patients is considered to evaluate its impact on cancer response to chemotherapy. We show that combining antiangiogenic and anticancer drugs improves the outcome of treatment and can decrease accumulation of the drug in normal tissue and enhance drug delivery to the tumor. Our results demonstrate that although both concurrent and neoadjuvant combination therapies can increase intratumoral drug exposure and therapeutic accuracy, neoadjuvant therapy surpasses this, especially against hyperglycemia. Our model provides mechanistic explanations for clinical observations of tumor progression and response to treatment and establishes a computational framework for exploring better treatment strategies.
Collapse
Affiliation(s)
| | - Bahar Firoozabadi
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Aliasghar Mozafari
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
17
|
Lee EK, Koo B, Hwangbo Y, Lee YJ, Baek JY, Cha YJ, Kim SY, Sim SH, Lee KS, Park IH, Lee H, Joo J, Go S, Heo SC, Moon MK. Incidence and disease course of new-onset diabetes mellitus in breast and colorectal cancer patients undergoing chemotherapy: A prospective multicenter cohort study. Diabetes Res Clin Pract 2021; 174:108751. [PMID: 33722701 DOI: 10.1016/j.diabres.2021.108751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/24/2021] [Accepted: 03/05/2021] [Indexed: 10/21/2022]
Abstract
AIMS To investigate the incidence of and risk factors for new-onset type 2 diabetes mellitus (DM) developed during chemotherapy that included steroids in cancer patients without DM. METHODS This multicenter, prospective, and observational cohort study enrolled 299 cancer patients without DM (aged > 18 years), planning 4-8 cycles of adjuvant chemotherapy. The endpoints were the incidence, remission rate, and independent determinants of new-onset DM during chemotherapy. RESULTS Between April 2015 and March 2018, 270 subjects with colorectal cancer or breast cancer (mean age, 51.0 years) completed the follow up (mean 39 months). Of whom, 17 subjects (6.3%) developed DM within a median time of 90 days (range, 17-359 days). Male sex (hazard ratio [HR], 15.839; 95% confidence interval [CI], 2.004-125.20) and impaired fasting glucose (IFG) at baseline (HR, 8.307; CI, 1.826-37.786) were independent risk factors. Six months after chemotherapy completion, 11/17 subjects (64.7%) experienced DM remission, associated with a significantly higher C-peptide level at baseline (C-peptide levels, 1.3 ng/mL in subjects with remission and 0.9 ng/mL in subjects without remission, age- and sex-adjusted P = 0.007). CONCLUSIONS DM incidence was 6.3% in patients who received chemotherapy with dexamethasone. Close monitoring for hyperglycemia is recommended, especially for men with IFG. TRIAL REGISTRATION ClinicalTrials.gov (NCT03062072).
Collapse
Affiliation(s)
- Eun Kyung Lee
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Thyroid Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Bokyung Koo
- Division of Endocrinology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yul Hwangbo
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Thyroid Cancer, National Cancer Center, Goyang, Republic of Korea
| | - You Jin Lee
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Thyroid Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Ji Yeon Baek
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Colorectal Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Yong Jun Cha
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Colorectal Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Sun Young Kim
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Colorectal Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Sung Hoon Sim
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Keun Seok Lee
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - In Hae Park
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea; Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Hyewon Lee
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Jungnam Joo
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, USA
| | - Sujeong Go
- Center for Thyroid Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Seung Chul Heo
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Min Kyong Moon
- Division of Endocrinology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|