1
|
Jensen CE, Deal AM, Srikanth S, Nyrop KA, Mitin N, LeBlanc MR, Muss HB, Rubinstein SM, Tuchman SA, Lichtman EI. Association of p16(INK4a), a biomarker of cellular senescence, with receipt of therapy and frailty status among adults with plasma cell disorders. J Geriatr Oncol 2024:102174. [PMID: 39706780 DOI: 10.1016/j.jgo.2024.102174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/13/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Affiliation(s)
- Christopher E Jensen
- University of North Carolina School of Medicine, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| | - Allison M Deal
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Shweta Srikanth
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kirsten A Nyrop
- University of North Carolina School of Medicine, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | | | - Matthew R LeBlanc
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA; University of North Carolina School of Nursing, Chapel Hill, NC, USA
| | - Hyman B Muss
- University of North Carolina School of Medicine, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Samuel M Rubinstein
- University of North Carolina School of Medicine, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Sascha A Tuchman
- University of North Carolina School of Medicine, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Eben I Lichtman
- University of North Carolina School of Medicine, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Wilson SR, Mitin N, Miller VLA, Smitherman AB, Carden MA. Adolescents and young adults with sickle cell disease exhibit accelerated aging with elevated T-cell p16 INK4a expression. Aging (Albany NY) 2024; 16:13225-13236. [PMID: 39546497 PMCID: PMC11719104 DOI: 10.18632/aging.206152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
People living with sickle cell disease (SCD) experience complications indicative of an accelerated aging phenotype typified by early decline in physical function and increased risk for age-related conditions. Cellular senescence, measured by expression of p16INK4a in peripheral T-lymphocytes, is recognized as one of the underlying contributors to organismal aging. To examine if cellular senescence is increased in SCD patients, we cross-sectionally measured and compared expression of p16 mRNA in peripheral blood T lymphocytes in 18 adolescents and young adults with SCD to 27 similarly aged individuals without SCD. Expression of p16 was dramatically higher in individuals with SCD vs. without SCD (10.1 vs. 8.7 log2 p16 units, respectively, p < 0.001) - a gap of 43 years in biological age - consistent with accelerated aging in the SCD population. Race was not associated with the increased p16 expression in the SCD group. These initial results suggest that individuals with SCD have a significantly higher cellular senescence burden which may contribute to premature aging, physiological decline, and excess morbidities. Additional longitudinal assessment and consideration for trials of senolytic therapies among individuals living with SCD and high p16 expression are warranted to improve their health span.
Collapse
Affiliation(s)
- Samuel R. Wilson
- Department of Medicine, Division of Hematology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalia Mitin
- Sapere Bio, Research Triangle Park, Raleigh-Durham-Chapel Hill, NC 27709, USA
| | - Vanessa L. Ayer Miller
- Department of Pharmaceutical and Clinical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Andrew B. Smitherman
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Department of Pharmaceutical and Clinical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Marcus A. Carden
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Cogent Biosciences, Waltham, MA 02451, USA
| |
Collapse
|
3
|
Carroll JE, Crespi CM, Cole S, Ganz PA, Petersen L, Bower JE. Transcriptomic markers of biological aging in breast cancer survivors: a longitudinal study. J Natl Cancer Inst 2024:djae201. [PMID: 39375195 DOI: 10.1093/jnci/djae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/09/2024] [Accepted: 08/15/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND The purpose of this study was to examine the impact of breast cancer therapy on biological aging as measured by expression of genes for cellular senescence (p16INK4a, SenMayo), DNA damage response, and proinflammatory senescence-associated secretory phenotype. METHODS This longitudinal, observational study evaluated women diagnosed with breast cancer (stage 0-III) prior to radiation therapy (RT) and/or chemotherapy (CT) and at repeated visits out to 2 years. Peripheral blood mononuclear cell gene expression was assessed using RNA sequencing on quality-verified RNA. Longitudinal data were analyzed using mixed linear models and a zero-inflated 2-part model. RESULTS Women (mean age = 55.5 years) receiving CT with or without RT (n = 73) had higher odds (odds ratio = 2.97, 95% confidence interval = 1.52 to 5.8) of having detectable p16INK4a following treatment compared with RT (n = 76) or surgery alone (n = 37). The proportion of women expressing 16INK4a over the follow-up period increased in all treatment groups (P < .001), with no interaction by treatment. All groups also increased over time in DNA damage response (P < .001), SenMayo (P < .001), and senescence-associated secretory phenotype (P < .001). Groups differed in the pattern of increase over time with statistically significant quadratic time by group differences for CT with or without RT compared with RT alone for DNA damage response (P = .05), SenMayo (P = .006), and the senescence-associated secretory phenotype (P = .02). CONCLUSIONS Results revealed activation of genes associated with biological aging in women with breast cancer from diagnosis through early survivorship, including DNA damage response, cell senescence, and the inflammatory secretome. Increases were evident across cancer treatments, although women receiving CT showed sustained increases, whereas RT exhibited slowing at later time points. Overall, findings suggest that women treated for breast cancer are aging within their immune cells.
Collapse
Affiliation(s)
- Judith E Carroll
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Catherine M Crespi
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Steve Cole
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Patricia A Ganz
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA
- Department of Health Policy and Management, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Laura Petersen
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Julienne E Bower
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Psychology, UCLA, Los Angeles, CA, USA
| |
Collapse
|
4
|
Ellis K, Koechlin H, Rudaz M, Gerido L, Hecht H, Jones C, Raji D, Northouse L, Katapodi M. Predictors and Interdependence of Quality of Life in a Random Sample of Long-Term Young Breast Cancer Survivors and Their Biological Relatives. Cancer Med 2024; 13:e70328. [PMID: 39470180 PMCID: PMC11519995 DOI: 10.1002/cam4.70328] [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: 05/20/2024] [Revised: 08/28/2024] [Accepted: 09/28/2024] [Indexed: 10/30/2024] Open
Abstract
PURPOSE Quality of life (QOL) among young breast cancer survivors (YBCS) is often worse than QOL of older breast cancer survivors or age-matched peers without a history of cancer. Families commonly support YBCS, particularly during treatment, but little is known about long-term YBCS and family member QOL. The purpose of this study was to identify demographic, clinical, and psychosocial predictors of physical and mental QOL in YBCS and biological relatives and investigate associations between their QOL (i.e., QOL interdependence). METHODS This secondary data analysis includes a random sample of long-term YBCS (≤ 45 years old at diagnosis) and up to two female relatives at baseline (post-treatment) and 18-month follow-up. The sample consists of 189 dyads (YBCS and one relative) and 121 triads (YBCS and two relatives). Actor-partner interdependence models (APIMs) were used to estimate the influence of YBCS's and relatives' demographic, clinical, and psychosocial factors on their own QOL (actor effects) and the other persons' QOL (partner effects). RESULTS For YBCS and relatives, QOL at the baseline was associated with their QOL at 18-months. YBCS's perceived cancer risk was associated with their own and relatives' QOL. Older relatives' physical QOL at baseline was associated with younger relatives' physical QOL at follow-up. Age, race, marital status, years since diagnosis, education, out-of-pocket costs of care, routine sources of care, income, family support, fear of recurrence, anxiety, and depression were also significant predictors of QOL. CONCLUSIONS Findings revealed independent and interdependent effects on QOL. These predictors point to potential targets of support for families. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT01612338.
Collapse
Affiliation(s)
- Katrina R. Ellis
- School of Social WorkUniversity of MichiganAnn ArborMichiganUSA
- School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
- Research Center for Group DynamicsInstitute for Social Research, University of MichiganAnn ArborMichiganUSA
| | | | - Marion Rudaz
- Department of Clinical ResearchUniversity of BaselBaselSwitzerland
| | | | - Hillary K. Hecht
- University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Carly Jones
- Michigan MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Dolapo Raji
- School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | | | - Maria Katapodi
- Department of Clinical ResearchUniversity of BaselBaselSwitzerland
| |
Collapse
|
5
|
El Jurdi N, Te HS, Cao Q, Napurski C, Wang S, Robinson A, Arora M, ElHusseini H, He F, Niedernhofer LJ, Thyagarajan B, Prizment A, Holtan S, Blaes AH, Yousefzadeh MJ. Frailty and pre-frailty associated with long-term diminished physical performance and quality of life in breast cancer and hematopoietic cell transplant survivors. Aging (Albany NY) 2024; 16:12432-12442. [PMID: 39330993 PMCID: PMC11466481 DOI: 10.18632/aging.206109] [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: 02/27/2024] [Accepted: 07/18/2024] [Indexed: 09/28/2024]
Abstract
Physical frailty as a sign of accelerated aging is not well characterized in breast cancer (BC) and hematopoietic cell transplant (HCT) survivors and its correlation with outcomes and quality of life (QOL) is not defined. We conducted a prospective study to determine the prevalence of frailty in adult BC and HCT survivors, examine its impact on QOL, and determine its association with p16INK4a, a molecular biomarker for biological aging. The study included 59 BC and 65 HCT survivors. Median age was 60 years (range 27-81), 68.5% were female and 49.2% were 18-59 vs. 51.8% ≥60 years old. A total of 71 (57.3%) were "fit" (frailty score 0) vs. 53 (42.7%) were pre-frailty/frail (frailty scores ≥1), and of the latter 17 (32.1%) were BC and 36 (67.9%) HCT patients. On multivariate analysis, patients >60 years were twice as likely to be frail (OR 2.04, 95% CI, 0.96-4.33; p=0.07), HCT were more likely to be frail compared to BC patients, and female HCT had 2.43 (95% CI, 0.92-6.40) and male HCT patients had 3.25 (95% CI, 1.37-7.72) times higher risk of frail; p=0.02. Frailty was associated with significant decline in QOL, measured by Medical Outcomes Study (MOS) Short Form 36 (SF-36) Physical Component Summary (PCS) and Mental Component Summary (MCS), and FACT (Functional Assessment of Cancer Therapy) scores. p16INK4a expression was higher in those who were frail, older than 60, and with higher expression in frail vs. fit patients who are 18-59 years. Our study highlights the high prevalence of frailty in survivors with detrimental effects on physical and overall wellbeing, and supports an association between frailty and the senescence marker p16INK4a.
Collapse
Affiliation(s)
- Najla El Jurdi
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
| | - Hok Sreng Te
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
| | - Qing Cao
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | - Char Napurski
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
| | - Shuo Wang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55454, USA
| | - Andre Robinson
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | | | - Heba ElHusseini
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
| | - Fiona He
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55454, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55454, USA
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55454, USA
| | - Shernan Holtan
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
| | - Anne Hudson Blaes
- Division of Hematology, Oncology, And Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55454, USA
| | - Matthew J. Yousefzadeh
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55454, USA
- Columbia Center for Translational Immunology and Columbia Center for Healthy Longevity, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
6
|
Sedrak MS, Sun CL, Bae M, Freedman RA, Magnuson A, O'Connor T, Moy B, Wildes TM, Klepin HD, Chapman AE, Tew WP, Dotan E, Fenton MA, Kim H, Katheria V, Muss HB, Cohen HJ, Gross CP, Ji J. Functional decline in older breast cancer survivors treated with and without chemotherapy and non-cancer controls: results from the Hurria Older PatiEnts (HOPE) prospective study. J Cancer Surviv 2024; 18:1131-1143. [PMID: 38678525 PMCID: PMC11324395 DOI: 10.1007/s11764-024-01594-3] [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: 10/03/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE This study aimed to assess whether physical functional decline in older women with early-stage breast cancer is driven by cancer, chemotherapy, or a combination of both. METHODS We prospectively sampled three groups of women aged ≥ 65: 444 with early-stage breast cancer receiving chemotherapy (BC Chemo), 98 with early-stage breast cancer not receiving chemotherapy (BC Control), and 100 non-cancer controls (NC Control). Physical function was assessed at two timepoints (T1 [baseline] and T2 [3, 4, or 6 months]) using the Physical Functioning Subscale (PF-10) of the RAND 36-item Short Form. The primary endpoint was the change in PF-10 scores from T1 to T2, analyzed continuously and dichotomously (Yes/No, with "yes" indicating a PF-10 decline > 10 points, i.e., a substantial and clinically meaningful difference). RESULTS Baseline PF-10 scores were similar across all groups. The BC Chemo group experienced a significant decline at T2, with a median change in PF-10 of -5 (interquartile range [IQR], -20, 0), while BC Control and NC Control groups showed a median change of 0 (IQR, -5, 5; p < 0.001). Over 30% of BC Chemo participants had a substantial decline in PF-10 vs. 8% in the BC Control and 5% in the NC Control groups (p < 0.001). CONCLUSION In this cohort of older adults with early-stage breast cancer, the combination of breast cancer and chemotherapy contributes to accelerated functional decline. Our findings reinforce the need to develop interventions aimed at preserving physical function, particularly during and after chemotherapy. IMPLICATIONS FOR CANCER SURVIVORS The high prevalence of accelerated functional decline in older women undergoing breast cancer chemotherapy underscores the urgency to develop interventions aimed at preserving physical function and improving health outcomes. CLINICAL TRIAL NCT01472094, Hurria Older PatiEnts (HOPE) with Breast Cancer Study.
Collapse
Affiliation(s)
- Mina S Sedrak
- Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
- Cancer & Aging Program, UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
- UCLA David Geffen School of Medicine, 650 Charles Young Drive South, Room A2-125 CHS, Los Angeles, CA, 90095-6900, USA.
| | - Can-Lan Sun
- Department of Supportive Care, City of Hope, Duarte, CA, USA
- Center for Cancer and Aging, City of Hope, Duarte, CA, USA
| | - Marie Bae
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA, USA
| | - Rachel A Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Allison Magnuson
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Tracey O'Connor
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Beverly Moy
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tanya M Wildes
- Department of Medicine, Medical Center/Nebraska Medicine, University of Nebraska, Omaha, NE, USA
| | - Heidi D Klepin
- Department of Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Andrew E Chapman
- Department of Medical Oncology, Sidney Kimmel Cancer Center/Jefferson Health, Philadelphia, PA, USA
| | - William P Tew
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Efrat Dotan
- Department of Hematology-Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Heeyoung Kim
- Department of Supportive Care, City of Hope, Duarte, CA, USA
- Center for Cancer and Aging, City of Hope, Duarte, CA, USA
| | - Vani Katheria
- Department of Supportive Care, City of Hope, Duarte, CA, USA
- Center for Cancer and Aging, City of Hope, Duarte, CA, USA
| | - Hyman B Muss
- Department of Medicine, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Harvey J Cohen
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Cary P Gross
- Cancer Outcomes Public Policy and Effectiveness Research (COPPER) Center, Yale School of Medicine, New Haven, CT, USA
| | - Jingran Ji
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA, USA
| |
Collapse
|
7
|
Rosko AE, Elsaid MI, Woyach J, Islam N, Lepola N, Urrutia J, Christian LM, Presley C, Mims A, Burd CE. Determining the relationship of p16 INK4a and additional molecular markers of aging with clinical frailty in hematologic malignancy. J Cancer Surviv 2024; 18:1168-1178. [PMID: 38678524 PMCID: PMC11324703 DOI: 10.1007/s11764-024-01591-6] [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: 10/03/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE Older adults with hematologic malignancies (HM) have unique challenges due to age and fitness. The primary aim of this pilot study was to benchmark the ability of multiple biomarkers of aging (p16, epigenetic clocks, T cell gene expression profiles, and T cell receptor excision circles (TREC) to identify frailty as measured by a clinical impairment index (I2) in patients with HM. METHODS 70 patients newly diagnosed with HM had peripheral blood T lymphocytes (PBTL) analyzed for p16INK4a expression using the OSU_Senescence Nanostring CodeSet. PBTL epigenetic age was measured using 7 epigenetic clocks, and TREC were quantified by qRT-PCR. A composite clinical impairment index (I2) was generated by combining values from 11 geriatric metrics (Independent Activities of Daily Living (iADL), physical health score, Short Physical Performance Battery (SPPB), Body Mass Index (BMI), Eastern Cooperative Oncology Group (ECOG) performance status, self-reported KPS, Blessed Orientation Memory Concentration (BOMC), polypharmacy, Mental Health Inventory (MHI)-17, Medical Outcomes Study (MOS) subscales). Clinical frailty was defined as a score of 7 or greater on the I2. RESULTS Age-adjusted p16INK4a was similar in newly diagnosed patients and healthy controls (p > 0.1). PBTL p16INK4a levels correlated positively with the Hannum [r = 0.35, 95% CI (0.09-0.75); p adj. = 0.04] and PhenoAge [r = 0.37, 95% CI (0.11-0.59); p adj. = 0.04] epigenetic clocks. The discrimination ability of the I2 model was calculated using the area under the receiver operating characteristic curve (AUC). After adjusting for chronologic age and disease group, baseline p16INK4a [AUC = 0.76, 95% CI (0.56-0.98); p = 0.01], Hannum [AUC = 0.70, 95% CI (0.54-0.85); p = 0.01], PhenoAge [AUC = 0.71, 95% CI (0.55-0.86); p = 0.01], and DunedinPACE [AUC = 0.73, 95% CI (0.57-0.88); p = < 0.01] measures showed the greatest potential to identify clinical frailty using the I2. CONCLUSIONS Our pilot data suggest that multiple blood-based aging biomarkers have potential to identify frailty in older adults with HM. IMPLICATIONS FOR CANCER SURVIVORS We developed the I2 index to quantify impairments across geriatric domains and discovered that PBTL p16, Hannum, PhenoAge, and DunedinPACE are promising indicators of frailty in HM.
Collapse
Affiliation(s)
- Ashley E Rosko
- Division of Hematology, The Ohio State University, Columbus, OH, USA.
- James Comprehensive Cancer Center, 300 West 10th Ave, Columbus, Ohio, 43210, United States.
| | - Mohamed I Elsaid
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Jennifer Woyach
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Nowshin Islam
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Noah Lepola
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Jazmin Urrutia
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Lisa M Christian
- Department of Psychiatry and Behavioral Health, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Carolyn Presley
- Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Alice Mims
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Christin E Burd
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
8
|
Christian LM, Kiecolt-Glaser JK, Cole SW, Burd CE, Madison AA, Wilson SJ, Rosko AE. Psychoneuroimmunology in multiple myeloma and autologous hematopoietic stem cell transplant: Opportunities for research among patients and caregivers. Brain Behav Immun 2024; 119:507-519. [PMID: 38643954 DOI: 10.1016/j.bbi.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024] Open
Abstract
Multiple myeloma (MM) is an incurable cancer and is the leading indication for autologous hematopoietic stem cell transplantation (HSCT). To be eligible for HSCT, a patient must have a caregiver, as caregivers play a central role in HSCT preparation and recovery. MM patients remain on treatment indefinitely, and thus patients and their caregivers face long-term challenges including the intensity of HSCT and perpetual therapy after transplant. Importantly, both patients and their caregivers show heightened depressive and anxiety symptoms, with dyadic correspondence evidenced and caregivers' distress often exceeding that of patients. An extensive psychoneuroimmunology (PNI) literature links distress with health via immune and neuroendocrine dysregulation as well as biological aging. However, data on PNI in the context of multiple myeloma - in patients or caregivers - are remarkably limited. Distress in MM patients has been associated with poorer outcomes including higher inflammation, greater one year post-HSCT hospital readmissions, and worse overall survival. Further, anxiety and depression are linked to biological aging and may contribute to the poor long-term health of both patients and caregivers. Because MM generally affects older adults, individual differences in biological aging may represent an important modifier of MM biology and HSCT treatment outcomes. There are a number of clinical scenarios in which biologically younger people could be prescribed more intensive therapies, with potential for greater benefit, by using a personalized cancer therapy approach based on the quantification of physiologic reserve. Further, despite considerable psychological demands, the effects of distress on health among MM caregivers is largely unexamined. Within this context, the current critical review highlights gaps in knowledge at the intersection of HSCT, inflammation, and biological aging in the context of MM. Research in this area hold promise for opportunities for novel and impactful psychoneuroimmunology (PNI) research to enhance health outcomes, quality of life, and longevity among both MM patients and their caregivers.
Collapse
Affiliation(s)
- Lisa M Christian
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA; The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Janice K Kiecolt-Glaser
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Steve W Cole
- Departments of Psychiatry and Biobehavioral Sciences and Medicine, Division of Hematology-Oncology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Christin E Burd
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA; Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Annelise A Madison
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Veteran's Affairs Boston Healthcare System, Boston, MA 02130, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Stephanie J Wilson
- Department of Psychology, Southern Methodist University, Dallas, TX 75206, USA
| | - Ashley E Rosko
- Division of Hematology, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
9
|
Justice JN, Leng XI, LeBrasseur NK, Tchkonia T, Kirkland JL, Mitin N, Liu Y, Kritchevsky SB, Nicklas BJ, Ding J. Caloric Restriction Intervention Alters Specific Circulating Biomarkers of the Senescence-Associated Secretome in Middle-Aged and Older Adults With Obesity and Prediabetes in an 18-Week Randomized Controlled Trial. J Gerontol A Biol Sci Med Sci 2024; 79:glad214. [PMID: 37738560 PMCID: PMC10733170 DOI: 10.1093/gerona/glad214] [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: 03/28/2023] [Indexed: 09/24/2023] Open
Abstract
Cellular senescence is a biological aging process that is exacerbated by obesity and leads to inflammation and age- and obesogenic-driven chronic diseases including type 2 diabetes. Caloric restriction (CR) may improve metabolic function in part by reducing cellular senescence and the pro-inflammatory senescence-associated phenotype (SASP). We conducted an ancillary investigation of an 18-week randomized controlled trial (RCT) of CR (n = 31) or Control (n = 27) in 58 middle-aged/older adults (57.6 ± 5.8 years; 75% Women) with obesity and prediabetes. We measured mRNA expression of select senescence and apoptosis genes in blood CD3 + T cells (qRT-PCR) and a panel of 25 plasma SASP proteins (Luminex/multiplex; ELISA). Participants randomized to CR lost -10.8 ± 0.9 kg (-11.3% ± 5.4%) over 18 weeks compared with +0.5 ± 0.9 kg (+0.03% ± 3.5%) in Control group. T-cell expression of senescence biomarkers, p16INK4a and p21CIP1/WAF1, and apoptosis markers, BCL2L1 and BAK1, was not different between CR and Control groups in age, race, and sex-adjusted mixed models (p > .05, all). Iterative principal axis factor analysis was used to develop composite SASP Factors, and the Factors comprising TNFRI, TNFRII, uPAR, MMP1, GDF15, OPN, Fas, and MPO were significantly altered with CR intervention (age, sex, race-adjusted mixed model time × treatment F = 4.17, p ≤ .05) and associated with the degree of weight loss (R2 = 0.12, p ≤ .05). Our study provides evidence from an RCT that specific circulating biomarkers of senescent cell burden are changed by CR in middle-aged and older adults with obesity and prediabetes. Future studies compare tissue and circulating levels of p16INK4a and pro-inflammatory SASP biomarkers in other populations, and interventions.
Collapse
Affiliation(s)
- Jamie N Justice
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Xiaoyan I Leng
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Natalia Mitin
- Sapere Bio, Triangle Research Park, North Carolina, USA
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Barbara J Nicklas
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jingzhong Ding
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
10
|
Rentscher KE, Bethea TN, Zhai W, Small BJ, Zhou X, Ahles TA, Ahn J, Breen EC, Cohen HJ, Extermann M, Graham DM, Jim HS, McDonald BC, Nakamura ZM, Patel SK, Root JC, Saykin AJ, Van Dyk K, Mandelblatt JS, Carroll JE. Epigenetic aging in older breast cancer survivors and noncancer controls: preliminary findings from the Thinking and Living with Cancer Study. Cancer 2023; 129:2741-2753. [PMID: 37259669 PMCID: PMC10659047 DOI: 10.1002/cncr.34818] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Cancer and its treatments may accelerate aging in survivors; however, research has not examined epigenetic markers of aging in longer term breast cancer survivors. This study examined whether older breast cancer survivors showed greater epigenetic aging than noncancer controls and whether epigenetic aging related to functional outcomes. METHODS Nonmetastatic breast cancer survivors (n = 89) enrolled prior to systemic therapy and frequency-matched controls (n = 101) ages 62 to 84 years provided two blood samples to derive epigenetic aging measures (Horvath, Extrinsic Epigenetic Age [EEA], PhenoAge, GrimAge, Dunedin Pace of Aging) and completed cognitive (Functional Assessment of Cancer Therapy-Cognitive Function) and physical (Medical Outcomes Study Short Form-12) function assessments at approximately 24 to 36 and 60 months after enrollment. Mixed-effects models tested survivor-control differences in epigenetic aging, adjusting for age and comorbidities; models for functional outcomes also adjusted for racial group, site, and cognitive reserve. RESULTS Survivors were 1.04 to 2.22 years biologically older than controls on Horvath, EEA, GrimAge, and DunedinPACE measures (p = .001-.04) at approximately 24 to 36 months after enrollment. Survivors exposed to chemotherapy were 1.97 to 2.71 years older (p = .001-.04), and among this group, an older EEA related to worse self-reported cognition (p = .047) relative to controls. An older epigenetic age related to worse physical function in all women (p < .001-.01). Survivors and controls showed similar epigenetic aging over time, but Black survivors showed accelerated aging over time relative to non-Hispanic White survivors. CONCLUSION Older breast cancer survivors, particularly those exposed to chemotherapy, showed greater epigenetic aging than controls that may relate to worse outcomes. If replicated, measurement of biological aging could complement geriatric assessments to guide cancer care for older women.
Collapse
Affiliation(s)
- Kelly E. Rentscher
- Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee WI
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| | - Traci N. Bethea
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Wanting Zhai
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Brent J. Small
- School of Aging Studies, University of South Florida, Tampa, FL
| | - Xingtao Zhou
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Tim A. Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Elizabeth C. Breen
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA
| | - Harvey Jay Cohen
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC
| | | | - Deena M.A. Graham
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | | | - Brenna C. McDonald
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine and Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | - Zev M. Nakamura
- Department of Psychiatry, University of North Carolina–Chapel Hill, Chapel Hill, NC
| | | | - James C. Root
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine and Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | - Kathleen Van Dyk
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| | | | - Judith E. Carroll
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
11
|
Ji J, Sun CL, Cohen HJ, Synold T, Muss H, Sedrak MS. Inflammation and Clinical Decline After Adjuvant Chemotherapy in Older Adults With Breast Cancer: Results From the Hurria Older Patients Prospective Study. J Clin Oncol 2023; 41:307-315. [PMID: 36126235 PMCID: PMC9839275 DOI: 10.1200/jco.22.01217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/18/2022] [Accepted: 08/08/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Older breast cancer survivors are at increased risk of clinical decline after adjuvant chemotherapy. This study aimed to evaluate whether inflammatory markers assessed before adjuvant chemotherapy are associated with chemotherapy-induced clinical decline in a population of fit older adults with breast cancer. METHODS In a prospective study of women age ≥ 65 years with stage I-III breast cancer treated with chemotherapy, we measured interleukin-6 (IL-6) and C-reactive protein (CRP) prechemotherapy (T1). We assessed frailty status, using a Deficit Accumulation Index (DAI; categorized as robust, prefrail, and frail), at T1 and postchemotherapy (T2). The population of interest was robust women at T1. The primary outcome was chemotherapy-induced decline in frailty status, defined as decline in DAI from robust (T1) to prefrail or frail (T2). Multivariable logistic regression was used to examine the association between inflammatory markers and the primary outcome, adjusted for sociodemographic and clinical characteristics. RESULTS Of the 295 robust women at T1, 76 (26%) experienced chemotherapy-induced decline in frailty status, among whom 66% had high IL-6, 63% had high CRP, and 46% had high IL-6 and CRP at T1. After adjusting for sociodemographic and clinical characteristics, women with high IL-6 and CRP had a > three-fold (odds ratio, 3.52; 95% CI, 1.55 to 8.01; P = .003) odds of chemotherapy-induced decline in frailty status compared with women with low IL-6 and CRP. CONCLUSION In this cohort of older women with early breast cancer who were clinically fit before chemotherapy initiation, high IL-6 and CRP prechemotherapy were associated with chemotherapy-induced decline in frailty status independent of sociodemographic and clinical risk factors. Further research is needed to examine whether inflammatory markers can inform more personalized approaches to treating older breast cancer survivors.
Collapse
Affiliation(s)
- Jingran Ji
- City of Hope National Medical Center, Duarte, CA
| | - Can-Lan Sun
- City of Hope National Medical Center, Duarte, CA
| | | | | | - Hyman Muss
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | | |
Collapse
|
12
|
Kammire MS, Deal AM, Damone EM, Rosen V, Nyrop KA, Mitin N, Muss HB. Does walking during chemotherapy impact p16 INK4a levels in women with early breast cancer. J Clin Lab Anal 2022; 36:e24753. [PMID: 36336905 PMCID: PMC9757016 DOI: 10.1002/jcla.24753] [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: 08/08/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Increased p16INK4a (p16) expression is directly related to cellular senescence and is a robust biomarker of aging in humans. Prior studies have shown that levels of p16 dramatically increase in breast cancer patients who have received adjuvant chemotherapy. This study investigated whether moderate physical activity during chemotherapy would attenuate the expected rise in p16 expression. METHODS Participants were women with Stage I-III breast cancer enrolled in a walking study for the duration of their chemotherapy (NCT02167932, NCT02328313, NCT03761706). Participants were asked to walk at least 30 min or 6200 steps/day following a structured walking program and to wear an activity tracker. p16 mRNA levels were measured in peripheral blood T-cells before chemotherapy initiation and at approximately 6 months after last chemotherapy treatment (mean 200 days, SD 40 days). RESULTS In total, 141 participants met inclusion criteria and 10% (n = 14) averaged > 6200 steps/day. There was no significant association of daily steps with change in p16 levels pre- to post-chemotherapy (Pearson correlation coefficient = 0.11, p = 0.17). After adjusting for age, stage, anthracycline-based chemotherapy, and baseline p16, the change in log2 p16 for each 1000 steps was estimated to be 0.03 (p = 0.35). Most participants were sedentary prior to chemotherapy and achieved modest levels of physical activity during treatment. CONCLUSION A self-guided walking program achieved only modest levels of physical activity and was unable to ameliorate chemotherapy-induced change in p16 levels in women undergoing chemotherapy for early-stage breast cancer. More structured and vigorous exercise programs should be tested for a more definitive exploration of their impact on post-chemotherapy p16 levels.
Collapse
Affiliation(s)
- Maria S. Kammire
- School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Allison M. Deal
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Emily M. Damone
- Gillings School of Global Public HealthUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Vanessa Rosen
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Kirsten A. Nyrop
- School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | | | - Hyman B. Muss
- School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
13
|
Mitin N, Nyrop KA, Strum SL, Knecht A, Carey LA, Reeder-Hayes KE, Claire Dees E, Jolly TA, Kimmick GG, Karuturi MS, Reinbolt RE, Speca JC, O'Hare EA, Muss HB. A biomarker of aging, p16, predicts peripheral neuropathy in women receiving adjuvant taxanes for breast cancer. NPJ Breast Cancer 2022; 8:103. [PMID: 36075910 PMCID: PMC9458644 DOI: 10.1038/s41523-022-00473-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Identifying patients at higher risk of chemotherapy-induced peripheral neuropathy (CIPN) is a major unmet need given its high incidence, persistence, and detrimental effect on quality of life. We determined if the expression of p16, a biomarker of aging and cellular senescence, predicts CIPN in a prospective, multi-center study of 152 participants enrolled between 2014 and 2018. Any women with newly diagnosed Stage I–III breast cancer scheduled to receive taxane-containing chemotherapy was eligible. The primary outcome was development of grade 2 or higher CIPN during chemotherapy graded by the clinician before each chemotherapy cycle (NCI-CTCAE v5 criteria). We measured p16 expression in peripheral blood T cells by qPCR before and at the end of chemotherapy. A multivariate model identified risk factors for CIPN and included taxane regimen type, p16Age Gap, a measure of discordance between chronological age and p16 expression, and p16 expression before chemotherapy. Participants with higher p16Age Gap—higher chronological age but lower p16 expression prior to chemotherapy - were at the highest risk. In addition, higher levels of p16 before treatment, regardless of patient age, conferred an increased risk of CIPN. Incidence of CIPN positively correlated with chemotherapy-induced increase in p16 expression, with the largest increase seen in participants with the lowest p16 expression before treatment. We have shown that p16 expression levels before treatment can identify patients at high risk for taxane-induced CIPN. If confirmed, p16 might help guide chemotherapy selection in early breast cancer.
Collapse
Affiliation(s)
| | - Kirsten A Nyrop
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Lisa A Carey
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine E Reeder-Hayes
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - E Claire Dees
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Trevor A Jolly
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Raquel E Reinbolt
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - JoEllen C Speca
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Erin A O'Hare
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hyman B Muss
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Shafqat S, Arana Chicas E, Shafqat A, Hashmi SK. The Achilles' heel of cancer survivors: fundamentals of accelerated cellular senescence. J Clin Invest 2022; 132:e158452. [PMID: 35775492 PMCID: PMC9246373 DOI: 10.1172/jci158452] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent improvements in cancer treatment have increased the lifespan of pediatric and adult cancer survivors. However, cancer treatments accelerate aging in survivors, which manifests clinically as the premature onset of chronic diseases, such as endocrinopathies, osteoporosis, cardiac dysfunction, subsequent cancers, and geriatric syndromes of frailty, among others. Therefore, cancer treatment-induced early aging accounts for significant morbidity, mortality, and health expenditures among cancer survivors. One major mechanism driving this accelerated aging is cellular senescence; cancer treatments induce cellular senescence in tumor cells and in normal, nontumor tissue, thereby helping mediate the onset of several chronic diseases. Studies on clinical monitoring and therapeutic targeting of cellular senescence have made considerable progress in recent years. Large-scale clinical trials are currently evaluating senotherapeutic drugs, which inhibit or eliminate senescent cells to ameliorate cancer treatment-related aging. In this article, we survey the recent literature on phenotypes and mechanisms of aging in cancer survivors and provide an up-to-date review of the major preclinical and translational evidence on cellular senescence as a mechanism of accelerated aging in cancer survivors, as well as insight into the potential of senotherapeutic drugs. However, only with time will the clinical effect of senotherapies on cancer survivors be visible.
Collapse
Affiliation(s)
| | - Evelyn Arana Chicas
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Shahrukh K. Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Clinical Affairs, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| |
Collapse
|
15
|
Carroll JE, Bower JE, Ganz PA. Cancer-related accelerated ageing and biobehavioural modifiers: a framework for research and clinical care. Nat Rev Clin Oncol 2022; 19:173-187. [PMID: 34873313 PMCID: PMC9974153 DOI: 10.1038/s41571-021-00580-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 12/15/2022]
Abstract
A growing body of evidence indicates that patients with cancer who receive cytotoxic treatments (such as chemotherapy or radiotherapy) have an increased risk of accelerated physical and cognitive ageing. Furthermore, accelerated biological ageing is a suspected driving force behind many of these observed effects. In this Review, we describe the mechanisms of biological ageing and how they apply to patients with cancer. We highlight the important role of specific behavioural factors, namely stress, sleep and lifestyle-related factors such as physical activity, weight management, diet and substance use, in the accelerated ageing of patients with cancer and cancer survivors. We also present a framework of how modifiable behaviours could operate to either increase the risk of accelerated ageing, provide protection, or promote resilience at both the biological level and in terms of patient-reported outcomes.
Collapse
Affiliation(s)
- Judith E Carroll
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behaviour, University of California, Los Angeles, CA, USA.
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
| | - Julienne E Bower
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behaviour, University of California, Los Angeles, CA, USA
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Patricia A Ganz
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Health Policy & Management, Fielding School of Public Health, University of California, Los Angeles, CA, USA
- Department of Medicine (Hematology-Oncology), David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Mandelblatt JS, Ahles TA, Lippman ME, Isaacs C, Adams-Campbell L, Saykin AJ, Cohen HJ, Carroll J. Applying a Life Course Biological Age Framework to Improving the Care of Individuals With Adult Cancers: Review and Research Recommendations. JAMA Oncol 2021; 7:1692-1699. [PMID: 34351358 PMCID: PMC8602673 DOI: 10.1001/jamaoncol.2021.1160] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance The practice of oncology will increasingly involve the care of a growing population of individuals with midlife and late-life cancers. Managing cancer in these individuals is complex, based on differences in biological age at diagnosis. Biological age is a measure of accumulated life course damage to biological systems, loss of reserve, and vulnerability to functional deterioration and death. Biological age is important because it affects the ability to manage the rigors of cancer therapy, survivors' function, and cancer progression. However, biological age is not always clinically apparent. This review presents a conceptual framework of life course biological aging, summarizes candidate measures, and describes a research agenda to facilitate clinical translation to oncology practice. Observations Midlife and late-life cancers are chronic diseases that may arise from cumulative patterns of biological aging occurring over the life course. Before diagnosis, each new patient was on a distinct course of biological aging related to past exposures, life experiences, genetics, and noncancer chronic disease. Cancer and its treatments may also be associated with biological aging. Several measures of biological age, including p16INK4a, epigenetic age, telomere length, and inflammatory and body composition markers, have been used in oncology research. One or more of these measures may be useful in cancer care, either alone or in combination with clinical history and geriatric assessments. However, further research will be needed before biological age assessment can be recommended in routine practice, including determination of situations in which knowledge about biological age would change treatment, ascertaining whether treatment effects on biological aging are short-lived or persistent, and testing interventions to modify biological age, decrease treatment toxic effects, and maintain functional abilities. Conclusions and Relevance Understanding differences in biological aging could ultimately allow clinicians to better personalize treatment and supportive care, develop tailored survivorship care plans, and prescribe preventive or ameliorative therapies and behaviors informed by aging mechanisms.
Collapse
Affiliation(s)
- Jeanne S Mandelblatt
- Department of Oncology, Cancer Prevention and Control Program, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC.,Department of Medicine, Georgetown University Medical Center, Washington, DC
| | - Tim A Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Marc E Lippman
- Department of Medicine, Georgetown University Medical Center, Washington, DC.,Department of Oncology, Breast Cancer Program, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Claudine Isaacs
- Department of Medicine, Georgetown University Medical Center, Washington, DC.,Department of Oncology, Breast Cancer Program, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Lucile Adams-Campbell
- Department of Oncology, Cancer Prevention and Control Program, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Andrew J Saykin
- Radiology and Imaging Sciences, Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research Center and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis
| | - Harvey J Cohen
- Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, North Carolina
| | - Judith Carroll
- UCLA Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Jonsson Comprehensive Cancer Center, and Cousins Center for Psychoneuroimmunology, Los Angeles, California
| |
Collapse
|
17
|
Russo M, Bono E, Ghigo A. The Interplay Between Autophagy and Senescence in Anthracycline Cardiotoxicity. Curr Heart Fail Rep 2021; 18:180-190. [PMID: 34081265 PMCID: PMC8342382 DOI: 10.1007/s11897-021-00519-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Doxorubicin (DOXO) is a highly effective chemotherapeutic drug employed for the treatment of a wide spectrum of cancers, spanning from solid tumours to haematopoietic malignancies. However, its clinical use is hampered by severe and dose-dependent cardiac side effects that ultimately lead to heart failure (HF). RECENT FINDINGS Mitochondrial dysfunction and oxidative stress are well-established mechanisms of DOXO-induced cardiotoxicity, although recent evidence suggests that deregulation of other biological processes, like autophagy, could be involved. It is increasingly recognized that autophagy deregulation is intimately interconnected with the initiation of detrimental cellular responses, including autosis and senescence, raising the possibility of using autophagy modulators as well as senolytics and senomorphics for preventing DOXO cardiotoxicity. This review aims at providing an overview of the signalling pathways that are common to autophagy and senescence, with a special focus on how the relationship between these two processes is deregulated in response to cardiotoxic treatments. Finally, we will discuss the potential therapeutic utility of drugs modulating autophagy and/or senescence for counteracting DOXO cardiotoxicity.
Collapse
Affiliation(s)
- Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Enrico Bono
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
18
|
Abdelgawad IY, Sadak KT, Lone DW, Dabour MS, Niedernhofer LJ, Zordoky BN. Molecular mechanisms and cardiovascular implications of cancer therapy-induced senescence. Pharmacol Ther 2021; 221:107751. [PMID: 33275998 PMCID: PMC8084867 DOI: 10.1016/j.pharmthera.2020.107751] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Cancer treatment has been associated with accelerated aging that can lead to early-onset health complications typically experienced by older populations. In particular, cancer survivors have an increased risk of developing premature cardiovascular complications. In the last two decades, cellular senescence has been proposed as an important mechanism of premature cardiovascular diseases. Cancer treatments, specifically anthracyclines and radiation, have been shown to induce senescence in different types of cardiovascular cells. Additionally, clinical studies identified increased systemic markers of senescence in cancer survivors. Preclinical research has demonstrated the potential of several approaches to mitigate cancer therapy-induced senescence. However, strategies to prevent and/or treat therapy-induced cardiovascular senescence have not yet been translated to the clinic. In this review, we will discuss how therapy-induced senescence can contribute to cardiovascular complications. Thereafter, we will summarize the current in vitro, in vivo, and clinical evidence regarding cancer therapy-induced cardiovascular senescence. Then, we will discuss interventional strategies that have the potential to protect against therapy-induced cardiovascular senescence. To conclude, we will highlight challenges and future research directions to mitigate therapy-induced cardiovascular senescence in cancer survivors.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Karim T Sadak
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA; University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Diana W Lone
- University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA
| | - Mohamed S Dabour
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| |
Collapse
|