1
|
Arjmand B, Mehran P, Badamchizadeh S, Alavi-Moghadam S, Arjmand R, Rezaei-Tavirani M, Aghayan HR, Larijani B, Vaezi M, Janbabaei G, Hajifathali A. The Role of Aging and Senescence in Bone Marrow Transplantation Outcome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40259169 DOI: 10.1007/5584_2025_861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Bone marrow transplantation is considered a cornerstone in the treatment of hematologic malignancies and blood disorders. While it may offer the possibility of a cure through the use of high-dose chemotherapy and radiation, outcomes are significantly impacted by biological and medical factors. Herein, aging is associated with reduced hematopoiesis, immune function, and overall regenerative capacity of tissues. Growth arrest, a crucial property of cellular senescence, inhibits bone marrow function, lowers immune surveillance in aged adults, and reduces the efficiency of bone marrow transplantation. The clinical course for older recipients is further complicated by the presence of prolonged immunosuppression, slower recovery, and higher complication rates, including life-threatening graft-versus-host disease. Accordingly, there is increasing interest in explaining how aging, cellular senescence, and transplant outcomes are interrelated. The current chapter outlines the mechanisms whereby aging and senescence contribute to the immunological dysregulation and poor bone marrow transplantation outcomes observed in elderly cancer patients. The authors' goal is to suggest therapeutic approaches that will enhance the quality of life and survival rates of elderly bone marrow transplant recipients.
Collapse
Affiliation(s)
- Babak Arjmand
- Hematology-Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Pouya Mehran
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Vaezi
- Hematology-Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Janbabaei
- Hematologic Malignancies Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Somay K, Albayrak Ö, Kızılırmak AB, Akan T, Üre ÜB, Akay OM, Ferhanoğlu B, Ateşoğlu EB. T cell subgroup analysis and T cell exhaustion after autologous stem cell transplantation in lymphoma patients. Transfus Apher Sci 2025; 64:104117. [PMID: 40222329 DOI: 10.1016/j.transci.2025.104117] [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: 05/21/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Autologous stem cell transplantation (ASCT) is a common treatment option for relapsed/refractory (R/R) lymphomas and it is considered standard of care as primary consolidation therapy for some types of Non-Hodgkin Lymphomas (NHL). Although ASCT benefits patients by allowing cytoreduction with intensive chemotherapy and reconstituting with stem cells, the effects of immunological changes in T cell subgroups after ASCT are still poorly understood. OBJECTIVES We evaluated changes in frequencies of T cell subsets and T cells expressing some of the exhaustion markers (such as LAG-3 and PD-1) from peripheral blood samples before and after ASCT to investigate bone marrow reconstruction and whether exhaustion predicts relapse. STUDY DESIGN Blood samples were collected on the day before conditioning and at the 1st, 3rd, and 6th months post-ASCT. Flow cytometry analysis was conducted to examine T cell subgroup composition and exhaustion markers, including PD-1 and LAG-3. Additionally, functional analysis was performed using assays for IFN-g and TNF-a production. Furthermore, a CSFE proliferation assay was utilized to assess proliferation capacity. RESULTS In our data set, dominant cells post-transplantation were memory cells, as the naïve cell population did not recover for 6 months. Both single and combined expressions of LAG-3 and PD-1 were found to be high before transplantation, and decreased after transplantation. However, LAG-3 and PD-1 expression increased in the 3rd and 6th month after transplantation respectively. These changes were more evident for the relapsed patients when compared to non-relapsed patients within 3 months follow-up time. Notably, the expression of inhibitory receptors in the relapsed patients was significantly higher at the first month post-transplantation. CD107a+ cytotoxic T lymphocytes (CTL), IFN-g+, TNF-a.+ CTL and T helper lymphocyte (THL) populations significantly decreased in relapsed patients 3rd month after transplantation. Decreased proliferation capacities of CTLs and THLs were also observed in these patients. CONCLUSION These results suggest that increased surface PD-1 and LAG-3 expressions along with functional decline after 3 months of ASCT can be used as prognostic data about the relapse status of transplant patients.
Collapse
Affiliation(s)
- Kayra Somay
- Department of Internal Medicine, Koç University Hospital, Istanbul, Turkey
| | - Özgür Albayrak
- Koç University Research Center for Translational Medicine (KUTTAM) Koç University Hospital, Istanbul, Turkey.
| | - Ali Burak Kızılırmak
- Koç University Research Center for Translational Medicine (KUTTAM) Koç University Hospital, Istanbul, Turkey.
| | - Tuba Akan
- Department of Hematology, Koç University Hospital, Istanbul, Turkey.
| | - Ümit Barbaros Üre
- Department of Hematology, Koç University Hospital, Istanbul, Turkey.
| | - Olga Meltem Akay
- Department of Hematology, Koç University Hospital, Istanbul, Turkey.
| | | | | |
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 2025; 117:312-321. [PMID: 39375195 PMCID: PMC11807438 DOI: 10.1093/jnci/djae201] [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: 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
|
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
|
5
|
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
|
6
|
Xiong J, Dong L, Lv Q, Yin Y, Zhao J, Ke Y, Wang S, Zhang W, Wu M. Targeting senescence-associated secretory phenotypes to remodel the tumour microenvironment and modulate tumour outcomes. Clin Transl Med 2024; 14:e1772. [PMID: 39270064 PMCID: PMC11398298 DOI: 10.1002/ctm2.1772] [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: 03/21/2024] [Revised: 06/17/2024] [Accepted: 07/08/2024] [Indexed: 09/15/2024] Open
Abstract
Tumour cell senescence can be induced by various factors, including DNA damage, inflammatory signals, genetic toxins, ionising radiation and nutrient metabolism. The senescence-associated secretory phenotype (SASP), secreted by senescent tumour cells, possesses the capacity to modulate various immune cells, including macrophages, T cells, natural killer cells and myeloid-derived suppressor cells, as well as vascular endothelial cells and fibroblasts within the tumour microenvironment (TME), and this modulation can result in either the promotion or suppression of tumorigenesis and progression. Exploring the impact of SASP on the TME could identify potential therapeutic targets, yet limited studies have dissected its functions. In this review, we delve into the causes and mechanisms of tumour cell senescence. We then concentrate on the influence of SASP on the tumour immune microenvironment, angiogenesis, extracellular matrix and the reprogramming of cancer stem cells, along with their associated tumour outcomes. Last, we present a comprehensive overview of the diverse array of senotherapeutics, highlighting their prospective advantages and challenge for the treatment of cancer patients. KEY POINTS: Senescence-associated secretory phenotype (SASP) secretion from senescent tumour cells significantly impacts cancer progression and biology. SASP is involved in regulating the remodelling of the tumour microenvironment, including immune microenvironment, vascular, extracellular matrix and cancer stem cells. Senotherapeutics, such as senolytic, senomorphic, nanotherapy and senolytic vaccines, hold promise for enhancing cancer treatment efficacy.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lu Dong
- The Second Clinical College of Wuhan University, Wuhan, China
| | - Qiongying Lv
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yutong Yin
- The First Clinical College of Wuhan University, Wuhan, China
| | - Jiahui Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Youning Ke
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Carballo‐Muñoz A, Lima G, Llorente L, Remolina‐Bonilla YA, Jaime‐Casas S, Otamendi‐Lopez A, Ortiz‐Guerra RA, Velazquez HE, Atisha‐Fregoso Y, Bourlon MT. Aging-related biomarkers in testicular cancer survivors after different oncologic treatments. Cancer Med 2024; 13:e70200. [PMID: 39300957 PMCID: PMC11413499 DOI: 10.1002/cam4.70200] [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: 11/24/2023] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE Testicular cancer survivors (TCS) exposed to chemotherapy have an increased expression of CDKN2A/p16INK4a and a lymphocyte phenotype associated with immunosenescence. We seek to define whether the immunosenescent phenotype is associated with chemotherapy. METHODS Case-control study of TCS, disease-free ≥3 months and stratified by primary treatment modality into orchiectomy only, chemotherapy, or bone marrow transplant (BMT). Each group was compared with age-matched healthy controls (HC). We measured the relative proportions of lymphocyte subpopulations using flow cytometry, levels of C-reactive protein, and relative expression of CDKN2A/p16INK4a quantified by qPCR. RESULTS We included 65 patients; 19 were treated with orchiectomy only, 35 received different doses of chemotherapy, and 11 underwent BMT. The chemotherapy and BMT groups had decreased naïve CD4 cells compared to HC. The chemotherapy group showed increased central and effector memory CD4 cells, as well as effector and terminally differentiated CD8 cells, compared to HC. Chemotherapy (chemotherapy 1.84 vs. HC 0.92; p < 0.01) and BMT (BMT 6.96 vs. HC 1.25; p < 0.005) groups had higher expression of CDKN2A/p16INK4a compared to HC. The orchiectomy group showed no significant difference with HC (orchiectomy 1.73 vs. HC 1.01; p = 0.17). CRP levels were higher in all groups when compared with HC; in the orchiectomy group, they were only marginally increased (chemotherapy 0.22 vs. HC 0.06; p < 0.01; BMT 0.26 vs. HC 0.06; p < 0.01; orchiectomy 0.09 vs. HC 0.07; p < 0.01). CONCLUSIONS Among TCS, only patients exposed to cytotoxic agents developed an immunosenescent phenotype. This finding supports the attribution of this alteration to the cytotoxic treatment.
Collapse
Affiliation(s)
- A. Carballo‐Muñoz
- Department of Hematology and OncologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| | - G. Lima
- Department of Inmunology and RheumatologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| | - L. Llorente
- Department of Inmunology and RheumatologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| | - Y. A. Remolina‐Bonilla
- Department of Hematology and OncologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| | - S. Jaime‐Casas
- Department of Hematology and OncologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| | - A. Otamendi‐Lopez
- Department of Hematology and OncologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| | - R. A. Ortiz‐Guerra
- Department of Hematology and OncologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| | - Hugo E. Velazquez
- Radiology DepartmentNational Institute of CardiologyMexico CityTlalpanMexico
| | - Y. Atisha‐Fregoso
- Institute of Molecular Medicine, Feinstein Institutes for Medical ResearchNew YorkNew YorkUSA
| | - M. T. Bourlon
- Department of Hematology and OncologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico CityTlalpanMexico
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Zhang W, Zhang K, Shi J, Qiu H, Kan C, Ma Y, Hou N, Han F, Sun X. The impact of the senescent microenvironment on tumorigenesis: Insights for cancer therapy. Aging Cell 2024; 23:e14182. [PMID: 38650467 PMCID: PMC11113271 DOI: 10.1111/acel.14182] [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: 01/17/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
The growing global burden of cancer, especially among people aged 60 years and over, has become a key public health issue. This trend suggests the need for a deeper understanding of the various cancer types in order to develop universally effective treatments. A prospective area of research involves elucidating the interplay between the senescent microenvironment and tumor genesis. Currently, most oncology research focuses on adulthood and tends to ignore the potential role of senescent individuals on tumor progression. Senescent cells produce a senescence-associated secretory phenotype (SASP) that has a dual role in the tumor microenvironment (TME). While SASP components can remodel the TME and thus hinder tumor cell proliferation, they can also promote tumorigenesis and progression via pro-inflammatory and pro-proliferative mechanisms. To address this gap, our review seeks to investigate the influence of senescent microenvironment changes on tumor development and their potential implications for cancer therapies.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Fang Han
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of EndocrinologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| |
Collapse
|
11
|
Englund DA, Jolliffe AM, Hanson GJ, Aversa Z, Zhang X, Jiang X, White TA, Zhang L, Monroe DG, Robbins PD, Niedernhofer LJ, Kamenecka TM, Khosla S, LeBrasseur NK. Senotherapeutic drug treatment ameliorates chemotherapy-induced cachexia. JCI Insight 2024; 9:e169512. [PMID: 38051584 PMCID: PMC10906225 DOI: 10.1172/jci.insight.169512] [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: 02/15/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023] Open
Abstract
Cachexia is a debilitating skeletal muscle wasting condition for which we currently lack effective treatments. In the context of cancer, certain chemotherapeutics cause DNA damage and cellular senescence. Senescent cells exhibit chronic activation of the transcription factor NF-κB, a known mediator of the proinflammatory senescence-associated secretory phenotype (SASP) and skeletal muscle atrophy. Thus, targeting NF-κB represents a logical therapeutic strategy to alleviate unintended consequences of genotoxic drugs. Herein, we show that treatment with the IKK/NF-κB inhibitor SR12343 during a course of chemotherapy reduces markers of cellular senescence and the SASP in liver, skeletal muscle, and circulation and, correspondingly, attenuates features of skeletal muscle pathology. Lastly, we demonstrate that SR12343 mitigates chemotherapy-induced reductions in body weight, lean mass, fat mass, and muscle strength. These findings support senescent cells as a promising druggable target to counteract the SASP and skeletal muscle wasting in the context of chemotherapy.
Collapse
Affiliation(s)
- Davis A. Englund
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Alyssa M. Jolliffe
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Gabriel J. Hanson
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Xinyi Jiang
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas A. White
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Lei Zhang
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - David G. Monroe
- Robert and Arlene Kogod Center on Aging, and
- Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, and
- Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
- Paul F. Glenn Center for the Biology of Aging at Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
12
|
Allemann MS, Lee P, Beer JH, Saeedi Saravi SS. Targeting the redox system for cardiovascular regeneration in aging. Aging Cell 2023; 22:e14020. [PMID: 37957823 PMCID: PMC10726899 DOI: 10.1111/acel.14020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiovascular aging presents a formidable challenge, as the aging process can lead to reduced cardiac function and heightened susceptibility to cardiovascular diseases. Consequently, there is an escalating, unmet medical need for innovative and effective cardiovascular regeneration strategies aimed at restoring and rejuvenating aging cardiovascular tissues. Altered redox homeostasis and the accumulation of oxidative damage play a pivotal role in detrimental changes to stem cell function and cellular senescence, hampering regenerative capacity in aged cardiovascular system. A mounting body of evidence underscores the significance of targeting redox machinery to restore stem cell self-renewal and enhance their differentiation potential into youthful cardiovascular lineages. Hence, the redox machinery holds promise as a target for optimizing cardiovascular regenerative therapies. In this context, we delve into the current understanding of redox homeostasis in regulating stem cell function and reprogramming processes that impact the regenerative potential of the cardiovascular system. Furthermore, we offer insights into the recent translational and clinical implications of redox-targeting compounds aimed at enhancing current regenerative therapies for aging cardiovascular tissues.
Collapse
Affiliation(s)
- Meret Sarah Allemann
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Pratintip Lee
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Jürg H. Beer
- Center for Molecular CardiologyUniversity of ZurichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital BadenBadenSwitzerland
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of CardiologyUniversity Hospital Zurich, University of ZurichSchlierenSwitzerland
| |
Collapse
|
13
|
Zhao B, Wu B, Feng N, Zhang X, Zhang X, Wei Y, Zhang W. Aging microenvironment and antitumor immunity for geriatric oncology: the landscape and future implications. J Hematol Oncol 2023; 16:28. [PMID: 36945046 PMCID: PMC10032017 DOI: 10.1186/s13045-023-01426-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
The tumor microenvironment (TME) has been extensively investigated; however, it is complex and remains unclear, especially in elderly patients. Senescence is a cellular response to a variety of stress signals, which is characterized by stable arrest of the cell cycle and major changes in cell morphology and physiology. To the best of our knowledge, senescence leads to consistent arrest of tumor cells and remodeling of the tumor-immune microenvironment (TIME) by activating a set of pleiotropic cytokines, chemokines, growth factors, and proteinases, which constitute the senescence-associated secretory phenotype (SASP). On the one hand, the SASP promotes antitumor immunity, which enhances treatment efficacy; on the other hand, the SASP increases immunosuppressive cell infiltration, including myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), M2 macrophages, and N2 neutrophils, contributing to TIME suppression. Therefore, a deeper understanding of the regulation of the SASP and components contributing to robust antitumor immunity in elderly individuals with different cancer types and the available therapies is necessary to control tumor cell senescence and provide greater clinical benefits to patients. In this review, we summarize the key biological functions mediated by cytokines and intercellular interactions and significant components of the TME landscape, which influence the immunotherapy response in geriatric oncology. Furthermore, we summarize recent advances in clinical practices targeting TME components and discuss potential senescent TME targets.
Collapse
Affiliation(s)
- Binghao Zhao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Departments of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100032, China
| | - Bo Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Nan Feng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiang Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| |
Collapse
|
14
|
Kruseova J, Zichova A, Eckschlager T. Premature aging in childhood cancer survivors. Oncol Lett 2022; 25:43. [PMID: 36644152 PMCID: PMC9811640 DOI: 10.3892/ol.2022.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/26/2022] [Indexed: 12/14/2022] Open
Abstract
Progress in medicine has increased the survival time of children suffering from cancer; >80% of patients survive for at least 5 years from the end of treatment. However, there are late effects of anticancer therapy, which accompany this success. Two-thirds of childhood cancer survivors (CCSs) have at least one late effect (any side effects or complications of anticancer treatment that appear months to years after the completion of treatment), e.g. endocrinopathies, cardiovascular diseases or subsequent cancers, and half of these late effects are serious or life threatening. These late consequences of childhood cancer treatment pose a serious health, social and economic problem. A common mechanism for developing a number of late effects is the onset of premature biological aging, which is associated with the early onset of chronic diseases and death. Cellular senescence in cancer survivors is caused by therapy that can induce chromosomal aberrations, mutations, telomere shortening, epigenetic alterations and mitochondrial dysfunctions. The mechanisms of accelerated aging in cancer survivors have not yet been fully clarified. The measurement of biological age in survivors can help improve the understanding of aging mechanisms and identify risk factors for premature aging. However, to the best of our knowledge, no single marker for the evaluation of biological or functional age is known, so it is therefore necessary to measure the consequences of anticancer treatment using complex assessments. The present review presents an overview of premature aging in CCSs and of the mechanisms involved in its development, focusing on the association of senescence and late effects.
Collapse
Affiliation(s)
- Jarmila Kruseova
- Department of Pediatric Hematology and Oncology, 2nd Medical Faculty, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic
| | - Andrea Zichova
- Department of Pediatric Hematology and Oncology, 2nd Medical Faculty, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic
| | - Tomas Eckschlager
- Department of Pediatric Hematology and Oncology, 2nd Medical Faculty, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic,Correspondence to: Professor Tomas Eckschlager, Department of Pediatric Hematology and Oncology, 2nd Medical Faculty, Charles University and University Hospital Motol, V Uvalu 84, 150 06 Prague, Czech Republic, E-mail:
| |
Collapse
|
15
|
Lázničková P, Bendíčková K, Kepák T, Frič J. Immunosenescence in Childhood Cancer Survivors and in Elderly: A Comparison and Implication for Risk Stratification. FRONTIERS IN AGING 2022; 2:708788. [PMID: 35822014 PMCID: PMC9261368 DOI: 10.3389/fragi.2021.708788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022]
Abstract
The population of childhood cancer survivors (CCS) has grown rapidly in recent decades. Although cured of their original malignancy, these individuals are at increased risk of serious late effects, including age-associated complications. An impaired immune system has been linked to the emergence of these conditions in the elderly and CCS, likely due to senescent immune cell phenotypes accompanied by low-grade inflammation, which in the elderly is known as "inflammaging." Whether these observations in the elderly and CCS are underpinned by similar mechanisms is unclear. If so, existing knowledge on immunosenescent phenotypes and inflammaging might potentially serve to benefit CCS. We summarize recent findings on the immune changes in CCS and the elderly, and highlight the similarities and identify areas for future research. Improving our understanding of the underlying mechanisms and immunosenescent markers of accelerated immune aging might help us to identify individuals at increased risk of serious health complications.
Collapse
Affiliation(s)
- Petra Lázničková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Tomáš Kepák
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Pediatric Oncology, University Hospital Brno, Brno, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
16
|
Guan L, Crasta KC, Maier AB. Assessment of cell cycle regulators in human peripheral blood cells as markers of cellular senescence. Ageing Res Rev 2022; 78:101634. [PMID: 35460888 DOI: 10.1016/j.arr.2022.101634] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022]
Abstract
Cellular senescence has gained increasing interest during recent years, particularly due to causal involvement in the aging process corroborated by multiple experimental findings. Indeed, cellular senescence considered to be one of the hallmarks of aging, is defined as a stable growth arrest predominantly mediated by cell cycle regulators p53, p21 and p16. Senescent cells have frequently been studied in the peripheral blood of humans due to its accessibility. This review summarizes ex vivo studies describing cell cycle regulators as markers of senescence in human peripheral blood cells, along with detection methodologies and associative studies examining demographic and clinical characteristics. The utility of techniques such as the quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), microarray, RNA sequencing and nCounter technologies for detection at the transcriptional level, along with Western blotting, enzyme-linked immunosorbent assay and flow cytometry at the translational level, will be brought up at salient points throughout this review. Notably, housekeeping genes or proteins serving as controls such as GAPDH and β-Actin, were found not to be stably expressed in some contexts. As such, optimization and validation of such genes during experimental design were recommended. In addition, the expression of cell cycle regulators was found to vary not only between different types of blood cells such as T cells and B cells but also between stages of cellular differentiation such as naïve T cells and highly differentiated T cells. On the other hand, the associations of the presence of cell cycle regulators with demographics (age, gender, ethnicity, and socioeconomic status), clinical characteristics (body mass index, specific diseases, disease-related parameters) and lifestyle vary in groups of participants. One envisions that increased understanding and insights into the assessment of cell cycle regulators as markers of senescence in human peripheral blood cells will help inform prognostication and clinical intervention in elderly individuals.
Collapse
Affiliation(s)
- Lihuan Guan
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia.
| | - Karen C Crasta
- Healthy Longevity Translational Researc h Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Physiology, National University of Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Agency for Science, Technology & Research (A⁎STAR), Institute of Molecular and Cell Biology (IMCB), Singapore.
| | - Andrea B Maier
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia; Healthy Longevity Translational Researc h Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore; Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, the Netherlands.
| |
Collapse
|
17
|
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: 46] [Impact Index Per Article: 15.3] [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
|
18
|
Ba R, Geffard E, Douillard V, Simon F, Mesnard L, Vince N, Gourraud PA, Limou S. Surfing the Big Data Wave: Omics Data Challenges in Transplantation. Transplantation 2022; 106:e114-e125. [PMID: 34889882 DOI: 10.1097/tp.0000000000003992] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In both research and care, patients, caregivers, and researchers are facing a leap forward in the quantity of data that are available for analysis and interpretation, marking the daunting "big data era." In the biomedical field, this quantitative shift refers mostly to the -omics that permit measuring and analyzing biological features of the same type as a whole. Omics studies have greatly impacted transplantation research and highlighted their potential to better understand transplant outcomes. Some studies have emphasized the contribution of omics in developing personalized therapies to avoid graft loss. However, integrating omics data remains challenging in terms of analytical processes. These data come from multiple sources. Consequently, they may contain biases and systematic errors that can be mistaken for relevant biological information. Normalization methods and batch effects have been developed to tackle issues related to data quality and homogeneity. In addition, imputation methods handle data missingness. Importantly, the transplantation field represents a unique analytical context as the biological statistical unit is the donor-recipient pair, which brings additional complexity to the omics analyses. Strategies such as combined risk scores between 2 genomes taking into account genetic ancestry are emerging to better understand graft mechanisms and refine biological interpretations. The future omics will be based on integrative biology, considering the analysis of the system as a whole and no longer the study of a single characteristic. In this review, we summarize omics studies advances in transplantation and address the most challenging analytical issues regarding these approaches.
Collapse
Affiliation(s)
- Rokhaya Ba
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Institute of Health and Medical Research, Centre de Recherche en Transplantation et Immunologie, UMR 1064, Institut de Transplantation Urologie-Néphrologie, Nantes, France
- Département Informatique et Mathématiques, Ecole Centrale de Nantes, Nantes, France
| | - Estelle Geffard
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Institute of Health and Medical Research, Centre de Recherche en Transplantation et Immunologie, UMR 1064, Institut de Transplantation Urologie-Néphrologie, Nantes, France
| | - Venceslas Douillard
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Institute of Health and Medical Research, Centre de Recherche en Transplantation et Immunologie, UMR 1064, Institut de Transplantation Urologie-Néphrologie, Nantes, France
| | - Françoise Simon
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Institute of Health and Medical Research, Centre de Recherche en Transplantation et Immunologie, UMR 1064, Institut de Transplantation Urologie-Néphrologie, Nantes, France
- Mount Sinai School of Medicine, New York, NY
| | - Laurent Mesnard
- Urgences Néphrologiques et Transplantation Rénale, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
- Sorbonne Université, Paris, France
| | - Nicolas Vince
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Institute of Health and Medical Research, Centre de Recherche en Transplantation et Immunologie, UMR 1064, Institut de Transplantation Urologie-Néphrologie, Nantes, France
| | - Pierre-Antoine Gourraud
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Institute of Health and Medical Research, Centre de Recherche en Transplantation et Immunologie, UMR 1064, Institut de Transplantation Urologie-Néphrologie, Nantes, France
| | - Sophie Limou
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Institute of Health and Medical Research, Centre de Recherche en Transplantation et Immunologie, UMR 1064, Institut de Transplantation Urologie-Néphrologie, Nantes, France
- Département Informatique et Mathématiques, Ecole Centrale de Nantes, Nantes, France
| |
Collapse
|
19
|
Maguire G. Stem cells part of the innate and adaptive immune systems as a therapeutic for Covid-19. Commun Integr Biol 2021; 14:186-198. [PMID: 34527167 PMCID: PMC8437473 DOI: 10.1080/19420889.2021.1965356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022] Open
Abstract
Some stem cell types not only release molecules that reduce viral replication, but also reduce the hypercytokinemia and inflammation induced by the immune system, and have been found to be part of the innate and adaptive immune systems. An important component of the stem cell's ability to ameliorate viral diseases, especially the complications post-clearance of the pathogen, is the ability of adult stem cells to reset the innate and adaptive immune systems from an inflammatory state to a repair state. Thus, the molecules released from certain stem cell types found to be safe and efficacious, may be an important new means for therapeutic development in Covid-19, especially for late-stage inflammation and tissue damage once the virus has cleared, particularly in the aged population.
Collapse
Affiliation(s)
- Greg Maguire
- Dept. of Preventative and Medicinal Chemistry, NeoGenesis Inc. And BioRegenerative Sciences Inc, San Diego, CA, USA
| |
Collapse
|
20
|
Mehata AK, Viswanadh MK, Priya V, Vikas, Muthu MS. Harnessing immunological targets for COVID-19 immunotherapy. Future Virol 2021. [PMID: 34447458 PMCID: PMC8375415 DOI: 10.2217/fvl-2021-0048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/16/2021] [Indexed: 12/22/2022]
Abstract
COVID-19 is an infectious and highly contagious disease caused by SARS-CoV-2. The immunotherapy strategy has a great potential to develop a permanent cure against COVID-19. Innate immune cells are in constant motion to scan molecular alteration to cells led by microbial infections throughout the body and helps in clearing invading viruses. Harnessing immunological targets for removing viral infection, generally based on the principle of enhancing the T-cell and protective immune responses. Currently-approved COVID-19 vaccines are mRNA encapsulated in liposomes that stimulate the host immune system to produce antibodies. Given the vital role of innate immunity, harnessing these immune responses opens up new hope for the generation of long-lasting and protective immunity against COVID-19.
Collapse
Affiliation(s)
- Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Matte Kasi Viswanadh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Vishnu Priya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Vikas
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| |
Collapse
|
21
|
Sedrak MS, Gilmore NJ, Carroll JE, Muss HB, Cohen HJ, Dale W. Measuring Biologic Resilience in Older Cancer Survivors. J Clin Oncol 2021; 39:2079-2089. [PMID: 34043454 PMCID: PMC8260901 DOI: 10.1200/jco.21.00245] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/24/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Affiliation(s)
| | | | - Judith E. Carroll
- University of California, Los Angeles, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, Jane & Terry Semel Institute for Neuroscience & Human Behavior, Department of Psychiatry & Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Los Angeles, CA
| | - Hyman B. Muss
- Department of Medicine and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | | | - William Dale
- City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
22
|
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
|
23
|
Lin RJ, Elias HK, van den Brink MRM. Immune Reconstitution in the Aging Host: Opportunities for Mechanism-Based Therapy in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2021; 12:674093. [PMID: 33953731 PMCID: PMC8089387 DOI: 10.3389/fimmu.2021.674093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Older patients with hematologic malignancies are increasingly considered for allogeneic hematopoietic cell transplantation with encouraging outcomes. While aging-related thymic dysfunction remains a major obstacle to optimal and timely immune reconstitution post- transplantation, recent accumulating evidence has suggested that various aging hallmarks such as cellular senescence, inflamm-aging, and hematopoietic stem cell exhaustion, could also impact immune reconstitution post-transplantation in both thymic-dependent and independent manner. Here we review molecular and cellular aspects of immune senescence and immune rejuvenation related to allogeneic hematopoietic cell transplantation among older patients and discuss potential strategies for mechanism-based therapeutic intervention.
Collapse
Affiliation(s)
- Richard J Lin
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Harold K Elias
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
24
|
Minnie SA, Hill GR. Autologous Stem Cell Transplantation for Myeloma: Cytoreduction or an Immunotherapy? Front Immunol 2021; 12:651288. [PMID: 33777050 PMCID: PMC7994609 DOI: 10.3389/fimmu.2021.651288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 02/19/2021] [Indexed: 12/22/2022] Open
Abstract
The incidence of multiple myeloma (MM), a bone marrow (BM) resident hematological malignancy, is increasing globally. The disease has substantial morbidity and mortality and remains largely incurable. Clinical studies show that autologous stem cell transplantation (ASCT) remains efficacious in eligible patients, providing a progression free survival (PFS) benefit beyond novel therapies alone. Conventionally, improved PFS after ASCT is attributed to cytoreduction from myeloablative chemotherapy. However, ASCT results in immune effects beyond cytoreduction, including inflammation, lymphodepletion, T cell priming via immunogenic cell death, and disruption of the tumor BM microenvironment. In fact, a small subset of patients achieve very long-term control of disease post-ASCT, akin to that seen in the context of immune-mediated graft-vs.-myeloma effects after allogeneic SCT. These clinical observations coupled with recent definitive studies in mice demonstrating that progression after ASCT represents immune escape as a consequence of T cell exhaustion, highlight the potential for new immunotherapy maintenance strategies to prevent myeloma progression following consolidation with ASCT.
Collapse
Affiliation(s)
- Simone A Minnie
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Division of Medical Oncology, University of Washington, Seattle, WA, United States
| |
Collapse
|
25
|
De Padova S, Urbini M, Schepisi G, Virga A, Meggiolaro E, Rossi L, Fabbri F, Bertelli T, Ulivi P, Ruffilli F, Casadei C, Gurioli G, Rosti G, Grassi L, De Giorgi U. Immunosenescence in Testicular Cancer Survivors: Potential Implications of Cancer Therapies and Psychological Distress. Front Oncol 2021; 10:564346. [PMID: 33520693 PMCID: PMC7844142 DOI: 10.3389/fonc.2020.564346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/23/2020] [Indexed: 01/13/2023] Open
Abstract
Testicular cancer (TC) is the most frequent solid tumor diagnosed in young adult males. Although it is a curable tumor, it is frequently associated with considerable short-term and long-term morbidity. Both biological and psychological stress experienced during cancer therapy may be responsible for stimulating molecular processes that induce premature aging and deterioration of immune system (immunosenescence) in TC survivors, leading to an increased susceptibility to infections, cancer, and autoimmune diseases. Immunosenescence is a remodeling of immune cell populations with inversion of the CD4:CD8 ratio, accumulation of highly differentiated memory cells, shrinkage of telomeres, shift of T-cell response to Th2 type, and release of pro-inflammatory signals. TC survivors exposed to chemotherapy show features of immunological aging, including an increase in memory T-cells (CD4+ and CD8+) and high expression of the senescence biomarker p16INK4a in CD3+ lymphocytes. However, the plethora of factors involved in the premature aging of TC survivors make the situation more complex if we also take into account the psychological stress and hormonal changes experienced by patients, as well as the high-dose chemotherapy and hematopoietic stem cell transplantation that some individuals may be required to undergo. The relatively young age and the long life expectancy of TC patients bear witness to the importance of improving quality of life and of alleviating long-term side-effects of cancer treatments. Within this context, the present review takes an in-depth look at the molecular mechanisms of immunosenescence, describing experimental evidence of cancer survivor aging and highlighting the interconnected relationship between the many factors modulating the aging of the immune system of TC survivors.
Collapse
Affiliation(s)
- Silvia De Padova
- Psycho-Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Milena Urbini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alessandra Virga
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Elena Meggiolaro
- Psycho-Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Lorena Rossi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Tatiana Bertelli
- Psycho-Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Federica Ruffilli
- Psycho-Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Gurioli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giovanni Rosti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Luigi Grassi
- Institute of Psychiatry, Department of Biomedical and Specialty Surgical Sciences, University of Ferrara and University Hospital Psychiatry Unit, Integrated Department of Mental Health S. Anna University Hospital and Health Authorities, Ferrara, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
26
|
Shachar SS, Deal AM, Reeder-Hayes KE, Nyrop KA, Mitin N, Anders CK, Carey LA, Dees EC, Jolly TA, Kimmick GG, Karuturi MS, Reinbolt RE, Speca JC, Muss HB. Effects of Breast Cancer Adjuvant Chemotherapy Regimens on Expression of the Aging Biomarker, p16INK4a. JNCI Cancer Spectr 2020; 4:pkaa082. [PMID: 33409457 PMCID: PMC7771421 DOI: 10.1093/jncics/pkaa082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/29/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Although chemotherapy saves lives, increasing evidence shows that chemotherapy accelerates aging. We previously demonstrated that mRNA expression of p16INK4a , a biomarker of senescence and molecular aging, increased early and dramatically after beginning adjuvant anthracycline-based regimens in early stage breast cancer patients. Here, we determined if changes in p16INK4a expression vary by chemotherapy regimen among early stage breast cancer patients. METHODS We conducted a study of stage I-III breast cancer patients receiving adjuvant or neoadjuvant chemotherapy. p16INK4a expression was analyzed prechemotherapy and postchemotherapy (median 6.2 months after the last chemotherapy) in peripheral blood T lymphocytes. Chemotherapy-induced change in p16INK4a expression was compared among regimens. All statistical tests were 2-sided. RESULTS In 146 women, chemotherapy was associated with a statistically significant increase in p16INK4a expression (accelerated aging of 17 years; P < .001). Anthracycline-based regimens were associated with the largest increases (accelerated aging of 23 to 26 years; P ≤ .008). Nonanthracycline-based regimens demonstrated a much smaller increase (accelerated aging of 9 to 11 years; P ≤ .15). In addition to the type of chemotherapy regimen, baseline p16INK4a levels, but not chronologic age or race, were also associated with the magnitude of increases in p16INK4a . Patients with lower p16INK4a levels at baseline were more likely to experience larger increases. CONCLUSIONS Our findings suggest that the aging effects of chemotherapy may be influenced by both chemotherapy type and the patient's baseline p16INK4a level. Measurement of p16INK4a expression is not currently available in the clinic, but nonanthracycline regimens offering similar efficacy as anthracycline regimens might be favored.
Collapse
Affiliation(s)
| | - Allison M Deal
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine E Reeder-Hayes
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kirsten A Nyrop
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - E Claire Dees
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Trevor A Jolly
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Raquel E Reinbolt
- Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | | | - Hyman B Muss
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
27
|
Smitherman AB, Wood WA, Mitin N, Ayer Miller VL, Deal AM, Davis IJ, Blatt J, Gold SH, Muss HB. Accelerated aging among childhood, adolescent, and young adult cancer survivors is evidenced by increased expression of p16 INK4a and frailty. Cancer 2020; 126:4975-4983. [PMID: 32830315 PMCID: PMC7607511 DOI: 10.1002/cncr.33112] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Cellular senescence, measured by expression of the cell cycle kinase inhibitor p16INK4a , may contribute to accelerated aging in survivors of childhood, adolescent, and young adult cancer. The authors measured peripheral blood T-lymphocyte p16INK4a expression among pediatric and young adult cancer survivors, hypothesizing that p16INK4a expression is higher after chemotherapy and among frail survivors. METHODS A cross-sectional cohort of young adult survivors and age-matched, cancer-free controls were assessed for p16INK4a expression and frailty. Newly diagnosed pediatric patients underwent prospective measurements of p16INK4a expression before and after cancer therapy. Frailty was measured with a modified Fried frailty phenotype evaluating sarcopenia, weakness, slowness, energy expenditure, and exhaustion. RESULTS The cross-sectional cohort enrolled 60 survivors and 29 age-matched controls with a median age of 21 years (range, 17-29 years). The prospective cohort enrolled 9 newly diagnosed patients (age range, 1-18 years). Expression of p16INK4a was higher among survivors compared with controls (9.6 vs 8.9 log2 p16 units; 2-sided P = .005, representing a 25-year age acceleration in survivors) and increased among newly diagnosed patients from matched pretreatment to posttreatment samples (7.3-8.9 log2 p16 units; 2-sided P = .002). Nine survivors (16%) were frail and had higher p16INK4a expression compared with robust survivors (10.5 [frail] vs 9.5 [robust] log2 p16 units; 2-sided P = .055), representing a 35-year age acceleration among frail survivors. CONCLUSIONS Chemotherapy is associated with increased cellular senescence and molecular age in pediatric and young adult cancer survivors. Frail survivors, compared with robust survivors, exhibit higher levels of p16INK4a , suggesting that cellular senescence may be associated with early aging in survivors.
Collapse
Affiliation(s)
- Andrew B. Smitherman
- Department of PediatricsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - William A. Wood
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Department of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | | | - Vanessa L. Ayer Miller
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Allison M. Deal
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Ian J. Davis
- Department of PediatricsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Julie Blatt
- Department of PediatricsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Stuart H. Gold
- Department of PediatricsUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Hyman B. Muss
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Department of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| |
Collapse
|
28
|
Burd CE, Peng J, Laskowski BF, Hollyfield JL, Zhang S, Fadda P, Yu L, Andridge RR, Kiecolt-Glaser JK. Association of Epigenetic Age and p16INK4a With Markers of T-Cell Composition in a Healthy Cohort. J Gerontol A Biol Sci Med Sci 2020; 75:2299-2303. [PMID: 32361724 PMCID: PMC7662168 DOI: 10.1093/gerona/glaa108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Indexed: 11/14/2022] Open
Abstract
How the measurement of aging biomarkers in peripheral blood T-lymphocytes (PBTLs) is influenced by cell composition is unclear. Here, we collected peripheral blood and isolated CD3+ PBTLs from 117 healthy couples between the ages of 21 and 72. Each sample was profiled for Horvath epigenetic clock (DNAm), p16INK4a expression, cytomegalovirus (CMV) seropositivity and 74 mRNA markers of PBTL subtype, differentiation, immune checkpoints, and cytokine production. Correlations between individual aging biomarkers (DNAm or p16INK4a) and PBTL mRNAs were corrected for chronological age, sex, and couple. DNAm measurements correlated with CMV seropositivity as well as PBTL mRNAs indicative of effector function (CD8A, EOMES, TBX21, GZMB), poor proliferative capacity (KLRG1, CD57), differentiation (CD45RO, CD45RA), and immune checkpoints (PDCD1, TIGIT, LAG3, CD160, CD244). In contrast, only three PBTL mRNAs, CD28, CD244, and p14ARF, showed a significant association with p16INK4a. p16INK4a expression also showed a weaker association with immunosenescent PBTL subsets than DNAm in flow cytometry analyses. These data suggest that PBTL composition has a greater influence on DNAm than p16INK4a and link accelerated epigenetic aging to immunosenescent phenotypes.
Collapse
Affiliation(s)
- Christin E Burd
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus
| | - Juan Peng
- Center for Biostatistics, Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus
| | - Bryon F Laskowski
- Department of Psychiatry and Behavioral Health, Institute for Behavioral Medicine, College of Medicine, The Ohio State University, Columbus
| | - Jennifer L Hollyfield
- Department of Psychiatry and Behavioral Health, Institute for Behavioral Medicine, College of Medicine, The Ohio State University, Columbus
| | - Suohui Zhang
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus
| | - Paolo Fadda
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus
| | - Lianbo Yu
- Center for Biostatistics, Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus
| | - Rebecca R Andridge
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus
| | - Janice K Kiecolt-Glaser
- Department of Psychiatry and Behavioral Health, Institute for Behavioral Medicine, College of Medicine, The Ohio State University, Columbus
| |
Collapse
|
29
|
Galloway JE, Holderbaum AM, Arya N, Zhang S, Bodnar MS, Norman R, Carson WE, Yu L, Kendra KL, Burd CE. Impact of age-related T cell dynamics on the identification of biomarkers predictive of immunotherapy discontinuation: a prospective cohort study. ACTA ACUST UNITED AC 2020; 1:58-70. [PMID: 34337428 DOI: 10.1002/aac2.12012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background The impact of biologic aging on immune checkpoint inhibitor (ICI) toxicity and efficacy is underexplored in metastatic melanoma (MM). In peripheral blood T-lymphocytes (PBTLs), biologic aging is characterized by changes in T-cell composition and cellular senescence. Whether indicators of PBTL biologic aging vary in MM patients or can be used to predict premature ICI discontinuation (pID) is unknown. Methods We prospectively collected PBTLs from 117 cancer-free controls and 46 MM patients scheduled to begin pembrolizumab or nivolumab monotherapy. 74 mRNAs indicative of T-cell subsets, activation, co-stimuation/inhibition and cellular senescence were measured by Nanostring. Relationships between each mRNA and chronologic age were assessed in patients and controls. Candidate biomarkers were identified by calculating the hazard ratio (HR) for pID in patients divided into low and high groups based on log-transformed mRNA levels or the magnitude by which each mRNA measurement deviated from the control trend (Δage). Area under the curve (AUC) analyses explored the ability of each biomarker to discriminate between patients with and without pID at 6 months and 1 year. Results Fifteen mRNAs correlated with chronologic age in controls, including markers of T-cell subsets, differentiation, cytokine production and co-stimulation/inhibition. None of these mRNAs remained correlated with age in patients. Median follow-up was 94.8 (1.6-195.7) weeks and 35 of 46 patients discontinued therapy (23 progression, 7 toxicity, 5 comorbidity/patient preference). Elevated pre-therapy CD8A (HR 2.2[1.1-4.9]), CD45RB (HR 2.9[1.4-5.8]) and TNFRSF14 (HR 2.2[1.1-4.5]) levels predicted pID independent of Δage-correction. CD3ε, CD27 and FOXO1 predicted pID only after Δage-correction (HR 2.5[1.3-5.1]; 3.7[1.8-7.8]; 2.1[1.1-4.3]). AUC analysis identified Δage-CD3ε and -CD27 as candidate predictors of pID (AUC=0.73; 0.75). Conclusions Correlations between transcriptional markers of PBTL composition and chronologic age are disrupted in MM. Correcting for normal, age-related trends in biomarker expression unveils new biomarker candidates predictive of ICI outcomes.
Collapse
Affiliation(s)
- Jason E Galloway
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Andrea M Holderbaum
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Namrata Arya
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Suohui Zhang
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Michael S Bodnar
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Ruthann Norman
- Department of Surgery, Division of Surgical Oncology, The Ohio State University College of Medicine, Columbus, OH 43210
| | - William E Carson
- Department of Surgery, Division of Surgical Oncology, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Lianbo Yu
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Kari L Kendra
- Medical Oncology Division, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| |
Collapse
|
30
|
Ness KK, Wogksch MD. Frailty and aging in cancer survivors. Transl Res 2020; 221:65-82. [PMID: 32360946 PMCID: PMC7321876 DOI: 10.1016/j.trsl.2020.03.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/01/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022]
Abstract
There are over 15 million survivors of cancer in the United States whose rates of frailty, an aging phenotype, range from just under 10% to over 80%. Frailty impacts not only disease survival but also long-term function and quality of life in children, adolescents, and in all adults diagnosed and/or treated for cancer. This review explains frailty as a construct and model of physiologic well-being. It also describes how frailty at diagnosis impacts cancer outcomes in adult populations and enumerates the prevalence of frailty in different populations of cancer survivors. Biological mechanisms responsible for aging and potentially for frailty among individuals with or who have been treated for cancer are discussed. Finally, promising pharmaceutical and lifestyle interventions designed to impact aging rather than a specific disease, tested in other populations, but likely applicable in cancer patients and survivors, are discussed.
Collapse
Affiliation(s)
- Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee.
| | - Matthew D Wogksch
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
31
|
Bruins WSC, Zweegman S, Mutis T, van de Donk NWCJ. Targeted Therapy With Immunoconjugates for Multiple Myeloma. Front Immunol 2020; 11:1155. [PMID: 32636838 PMCID: PMC7316960 DOI: 10.3389/fimmu.2020.01155] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
The introduction of proteasome inhibitors (PI) and immunomodulatory drugs (IMiD) has markedly increased the survival of multiple myeloma (MM) patients. Also, the unconjugated monoclonal antibodies (mAb) daratumumab (anti-CD38) and elotuzumab (anti-SLAMF7) have revolutionized MM treatment given their clinical efficacy and safety, illustrating the potential of targeted immunotherapy as a powerful treatment strategy for MM. Nonetheless, most patients eventually develop PI-, IMiD-, and mAb-refractory disease because of the selection of resistant MM clones, which associates with a poor prognosis. Accordingly, these patients remain in urgent need of new therapies with novel mechanisms of action. In this respect, mAbs or mAb fragments can also be utilized as carriers of potent effector moieties to specifically target surface antigens on cells of interest. Such immunoconjugates have the potential to exert anti-MM activity in heavily pretreated patients due to their distinct and pleiotropic mechanisms of action. In addition, the fusion of highly cytotoxic compounds to mAbs decreases the off-target toxicity, thereby improving the therapeutic window. According to the effector moiety, immunoconjugates are classified into antibody-drug conjugates, immunotoxins, immunocytokines, or radioimmunoconjugates. This review will focus on the mechanisms of action, safety and efficacy of several promising immunoconjugates that are under investigation in preclinical and/or clinical MM studies. We will also include a discussion on combination therapy with immunoconjugates, resistance mechanisms, and future developments.
Collapse
Affiliation(s)
- Wassilis S C Bruins
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tuna Mutis
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
32
|
The safety of a therapeutic product composed of a combination of stem cell released molecules from adipose mesenchymal stem cells and fibroblasts. Future Sci OA 2020; 6:FSO592. [PMID: 32802397 PMCID: PMC7421258 DOI: 10.2144/fsoa-2020-0027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: We sought to determine the safety profile of a therapeutic candidate composed of the released molecules from a combination of human adipose-derived mesenchymal stem cells and fibroblasts. Although stem cells, their progenitor cells and the molecules that are released from these cells have some demonstrated therapeutic value, much more needs to learn about the efficacy, mechanism of action and the safety profiles of these stem cell-based therapeutics. Methods: A number of cellular, in vitro, in vivo and human studies were performed to analyze cellular, tissue and systemic safety profiles of the combinatorial product. Results: At the levels tested in this study, ranging from demonstrated therapeutic doses to supratherapeutic doses, the combinatorial product demonstrated an excellent safety profile in all in vitro, cellular, tissue and systemic studies. Conclusions: We found evidence that a therapeutic candidate composed of the molecules released from human adipose-derived mesenchymal stem cells and human fibroblasts has an excellent safety profile, and that the product warrants further studies for safety and efficacy where dosing may include topical application, injection and oral application. Stem cell transplants have demonstrated life-saving capabilities for some diseases, and the molecules released from stem cells are currently in therapeutic development for a number of conditions. Stem cell science is a relatively new science and is in need of a better understanding of mechanisms of action and acute and long-term safety profiles. Here, we performed a number of safety tests for stem cell released molecules from a combination of adipose-derived mesenchymal stem cells and fibroblasts that have demonstrated efficacy in a number of conditions. Using in vitro, in vivo and skin sensitivity studies in humans, the stem cell therapeutic was found to have an excellent safety profile when tested for toxicity, mutagenicity, tumorigenesis, ocular toxicity, inflammation and irritation.
Collapse
|
33
|
Maguire G. Better preventing and mitigating the effects of Covid-19. Future Sci OA 2020; 6:FSO586. [PMID: 32685190 PMCID: PMC7238752 DOI: 10.2144/fsoa-2020-0051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/29/2020] [Indexed: 01/08/2023] Open
Abstract
Currently, there are no proven medical treatments against SARS-CoV-2, the virus responsible for Covid-19. In addition to the all important public health measures needed to prevent the spread of this disease, a number of strategies related to our exposome are recommended herein, to better prevent and mitigate the effects of a SARS-CoV-2 infection through enhancement of our immune system and reduction of inflammation.
Collapse
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences Inc., NeoGenesis Inc., San Diego, CA 94704, USA
- The California Physiological Society, Berkeley, CA 94704, USA
| |
Collapse
|
34
|
Abstract
Most cancers arise in individuals over the age of 60. As the world population is living longer and reaching older ages, cancer is becoming a substantial public health problem. It is estimated that, by 2050, more than 20% of the world's population will be over the age of 60 - the economic, healthcare and financial burdens this may place on society are far from trivial. In this Review, we address the role of the ageing microenvironment in the promotion of tumour progression. Specifically, we discuss the cellular and molecular changes in non-cancerous cells during ageing, and how these may contribute towards a tumour permissive microenvironment; these changes encompass biophysical alterations in the extracellular matrix, changes in secreted factors and changes in the immune system. We also discuss the contribution of these changes to responses to cancer therapy as ageing predicts outcomes of therapy, including survival. Yet, in preclinical studies, the contribution of the aged microenvironment to therapy response is largely ignored, with most studies designed in 8-week-old mice rather than older mice that reflect an age appropriate to the disease being modelled. This may explain, in part, the failure of many successful preclinical therapies upon their translation to the clinic. Overall, the intention of this Review is to provide an overview of the interplay that occurs between ageing cell types in the microenvironment and cancer cells and how this is likely to impact tumour metastasis and therapy response.
Collapse
Affiliation(s)
- Mitchell Fane
- The Wistar Institute, Immunology, Microenvironment and Metastasis Program, Philadelphia, PA, USA.
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA.
| | - Ashani T Weeraratna
- The Wistar Institute, Immunology, Microenvironment and Metastasis Program, Philadelphia, PA, USA.
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
35
|
Weiss JM, Pennell N, Deal AM, Morgensztern D, Bradford DS, Crane J, West HJ, Lee C, Pecot C, Stevenson JP, Irvin W, Socinski M, Stinchcombe T, Villaruz LC, Muss HB. Nab-paclitaxel in older patients with non-small cell lung cancer who have developed disease progression after platinum-based doublet chemotherapy. Cancer 2020; 126:1060-1067. [PMID: 31943168 DOI: 10.1002/cncr.32573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND The selection of later-line treatment for older patients with AJCC (version 7) stage IV non-small cell lung cancer (NSCLC) remains controversial. Nanoparticle albumin-bound (nab)-paclitaxel is approved with carboplatin for the first-line treatment of patients with NSCLC and subgroup analysis of phase 3 data has suggested superior survival in older patients. METHODS The authors conducted a phase 2 study of nab-paclitaxel in 42 patients aged ≥70 years who had been treated previously with a platinum doublet regimen; patients also could have received a PD-1 inhibitor. The primary endpoint of the current study was grade 3 to 5 toxicity (according to the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). In addition to response rate, progression-free survival (PFS), and overall survival (OS), geriatric assessments also were performed before and during treatment, associations between baseline sarcopenia and outcomes were explored, and changes in T lymphocyte p16 before and during treatment were measured. The authors also performed a retrospective subgroup analysis of 19 older patients who were treated with nab-paclitaxel as part of a larger, randomized, phase 2 study; data were not combined. RESULTS The rate of grade 3 to 5 toxicities was 33.7%. The most common grade 3 to 5 toxicities were decreased white blood cell count (11.9%), neutropenia (9.5%), and fatigue (11.9%). The response rate was 34.2% (2.6% complete response rate and 31.6% partial response rate). The median PFS was 5.2 months and the median OS was 9.3 months. Adverse prognostic factors were common: 42% of patients were frail and 39% of patients were prefrail, whereas 21% had an Eastern Cooperative Oncology Group performance status of 2 and 27% were sarcopenic. Only frailty was found to be predictive of inferior survival. A subgroup analysis of 19 older patients treated with nab-paclitaxel alone in a prior trial demonstrated a response rate of 15.8%, a PFS of 4.2 months, and an OS of 13.6 months. CONCLUSIONS Fit and prefrail older patients with stage IV NSCLC should be considered for treatment with nab-paclitaxel after disease progression with doublet chemotherapy.
Collapse
Affiliation(s)
- Jared M Weiss
- Department of Hematology and Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nathan Pennell
- Hematology and Medical Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Allison M Deal
- Department of Hematology and Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Daniel Morgensztern
- Oncology Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | | | - Jeffrey Crane
- Rex Hematology Oncology Associates, Raleigh-Durham, North Carolina
| | - Howard Jack West
- Department of Medical Oncology, Swedish Cancer Institute, Seattle, Washington
| | - Carrie Lee
- Department of Hematology and Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chad Pecot
- Department of Hematology and Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | - Tom Stinchcombe
- Division of Medical Oncology, Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| | - Liza C Villaruz
- Division of Hematology/Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Hyman B Muss
- Department of Hematology and Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
36
|
Henneghan A, Rao V, Harrison RA, Karuturi M, Blayney DW, Palesh O, Kesler SR. Cortical Brain Age from Pre-treatment to Post-chemotherapy in Patients with Breast Cancer. Neurotox Res 2020; 37:788-799. [PMID: 31900898 DOI: 10.1007/s12640-019-00158-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/18/2019] [Accepted: 12/22/2019] [Indexed: 12/12/2022]
Abstract
Chemotherapy-related cognitive impairment and associated brain changes may reflect accelerated brain aging; however, empirical evidence for this theory is limited. The purpose of this study was to measure brain aging in newly diagnosed patients with breast cancer treated with chemotherapy (n = 43) and compare its longitudinal change to that of controls (n = 50). Brain age indices, derived from cortical measures, were compared between women with breast cancer and matched healthy controls across 3 timepoints (time 1: pre-surgery, time 2: 1 month following chemotherapy completion, and time 3: 1-year post-chemotherapy). The breast cancer group showed a significant decrease in cortical thickness across the 3 timepoints (p < .001) and a trend towards significant increase in predicted brain age especially from pre-treatment (time 1) to post-chemotherapy (time 2) compared to controls (p = 0.08). Greater increase in predicted brain age was related to several clinical factors (HER-2 status, surgery type, and history of neoadjuvant chemotherapy) and greater decrease in cortical thickness was associated with greater decrease in performance on a verbal learning task from time 1 to time 3 (r = - 0.48, p < .01). This study demonstrated evidence of increased cortical brain aging in middle-aged patients with breast cancer following chemotherapy treatment that was associated with decreased verbal memory performance.
Collapse
Affiliation(s)
- Ashley Henneghan
- University of Texas at Austin School of Nursing, 1710 Red River St, Austin, TX, 78701, USA. .,Department of Oncology, Dell Medical School, University of Texas at Austin, 1601 Trinity St, Austin, TX, USA.
| | - Vikram Rao
- University of Texas at Austin School of Nursing, 1710 Red River St, Austin, TX, 78701, USA
| | - Rebecca A Harrison
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 431, Houston, TX, 77030, USA
| | - Meghan Karuturi
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Unit 1354, 1155 Pressler St., Houston, TX, 77030, USA
| | - Douglas W Blayney
- Division of Medical Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Oxana Palesh
- Department of Psychiatry and Behavioral Sciences, Stanford University Cancer Institute, Stanford University, 401 Quarry Road, Office 2318, Stanford, CA, 94305, USA
| | - Shelli R Kesler
- University of Texas at Austin School of Nursing, 1710 Red River St, Austin, TX, 78701, USA.,Department of Oncology, Dell Medical School, University of Texas at Austin, 1601 Trinity St, Austin, TX, USA
| |
Collapse
|
37
|
The Impact of Type II Diabetes on Tongue Dysplasia and p16-Related Aging Process: An Experimental Study. Anal Cell Pathol (Amst) 2019; 2019:3563215. [PMID: 31687322 PMCID: PMC6800967 DOI: 10.1155/2019/3563215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/25/2019] [Accepted: 09/09/2019] [Indexed: 11/24/2022] Open
Abstract
Objective To evaluate the effect of streptozotocin-induced experimental diabetes mellitus on p16, p53, Ki67, and Bcl2 expressions and histopathological changes in the tongue of the rats. Material and Methods Twenty-two adult female Sprague-Dawley rats were used. The rats were randomly divided into 2 groups (n = 14) as control (C) (n = 8) and diabetic (DM) (n = 6). The rats in the DM group were given streptozotocin as a single intraperitoneal dose for induction of diabetes. Histopathological and immunohistochemical evaluations of formalin-fixed and paraffin-embedded tissue sections of the tongue were used. Results Significant differences were observed between the DM group and the control group in terms of epithelial thickness, length of filiform papillae, and width of filiform papillae (p = 0.005, p = 0.001, and p = 0.006, respectively). There was no significant difference between the groups in terms of mononuclear inflammatory cell infiltration, capillary proliferation, and dysplasia (p = 0.204, p = 0.244, and p = 0.204, respectively). As a result of immunohistochemical studies, no significant difference was found between the groups in terms of p53, Ki67, and Bcl-2 expressions (p = 0.588, p = 0.662, and p = 0.686, respectively). A significant difference was found between the groups when p16 expression was evaluated (p = 0.006). Conclusions In our study, streptozotocin-induced experimental diabetes mellitus induced p16 expression but did not show any difference in p53, Bcl-2, and Ki67 levels. It should be considered in the studies that the pathological changes at the early stages of the relationship between DM and oral cancer may be related to p16 expression; however, it may also be linked with p16-related aging process.
Collapse
|
38
|
Maguire G. Transplanted stem cells survive a long time: do they make you sick? J R Soc Med 2019; 112:412-414. [PMID: 31526208 DOI: 10.1177/0141076819851657] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences Inc., San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
39
|
Lucas F, Pennell M, Huang Y, Benson DM, Efebera YA, Chaudhry M, Hughes T, Woyach JA, Byrd JC, Zhang S, Jones D, Guan X, Burd CE, Rosko AE. T Cell Transcriptional Profiling and Immunophenotyping Uncover LAG3 as a Potential Significant Target of Immune Modulation in Multiple Myeloma. Biol Blood Marrow Transplant 2019; 26:7-15. [PMID: 31445183 DOI: 10.1016/j.bbmt.2019.08.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/16/2019] [Accepted: 08/11/2019] [Indexed: 12/20/2022]
Abstract
Autologous stem cell transplant (ASCT) is the standard of care for patients with multiple myeloma (MM). The clinical significance of peripheral blood T lymphocyte (PBTL) immunologic changes associated with ASCT is poorly understood. Here we evaluated T cell transcriptional messenger RNA profiles and immunophenotypes to correlate immunologic senescence, exhaustion, and anergy with clinical endpoints in a cohort of patients with MM undergoing ASCT. ASCT induced global transcriptional T cell changes and altered molecular levels of markers of T cell subtypes, T cell activation, and exhaustion. These included reduced CD4/CD8 ratio, skewing toward the Th1 subset, reduced expression of costimulatory receptors CD27 and CD28, heightened T cell activation, and increased expression of immune modulatory molecules LAG3 and PD1. Multicolor flow cytometry experiments confirmed altered circulating CD4 and CD8 subsets and skewing toward differentiated effector cells. Moreover, ASCT promoted an exhausted immunophenotype in CD3+CD4+ subsets and a senescent immunophenotype in CD3+CD8+ subsets. Subset-specific altered expression was also seen for surface molecules with immunomodulatory function. ASCT affected soluble levels of molecules with immunomodulatory function by increasing plasma HVEM and TIM3. High molecular LAG3 level was associated with inferior event-free survival post-ASCT (hazard ratio = 5.44; confidence interval, 1.92 to 15.46; P = .001; adjusted P [controlling for false discovery rate] = .038). Using a comprehensive evaluation of PBTLs on a molecular and phenotypic level, we have identified that ASCT induces global T cell alterations with CD4 and CD8 subset-specific changes. Moreover, LAG3 emerged as an early biomarker of adverse events post-ASCT. These findings will support the development of treatment strategies targeting immune defects in MM to augment or restore T cell responses.
Collapse
Affiliation(s)
- Fabienne Lucas
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Michael Pennell
- College of Public Health, Division of Biostatistics, The Ohio State University, Columbus, Ohio
| | - Ying Huang
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Don M Benson
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Yvonne A Efebera
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Maria Chaudhry
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Tiffany Hughes
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | | | - John C Byrd
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Suohui Zhang
- Department of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Desiree Jones
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Xiangnan Guan
- Department of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Christin E Burd
- Department of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Ashley E Rosko
- Division of Hematology, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
40
|
Yousefzadeh MJ, Melos KI, Angelini L, Burd CE, Robbins PD, Niedernhofer LJ. Mouse Models of Accelerated Cellular Senescence. Methods Mol Biol 2019; 1896:203-230. [PMID: 30474850 DOI: 10.1007/978-1-4939-8931-7_17] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Senescent cells accumulate in multiple tissues as virtually all vertebrate organisms age. Senescence is a highly conserved response to many forms of cellular stress intended to block the propagation of damaged cells. Senescent cells have been demonstrated to play a causal role in aging via their senescence-associated secretory phenotype and by impeding tissue regeneration. Depletion of senescent cells either through genetic or pharmacologic methods has been demonstrated to extend murine lifespan and delay the onset of age-related diseases. Measuring the burden and location of senescent cells in vivo remains challenging, as there is no marker unique to senescent cells. Here, we describe multiple methods to detect the presence and extent of cellular senescence in preclinical models, with a special emphasis on murine models of accelerated aging that exhibit a more rapid onset of cellular senescence.
Collapse
Affiliation(s)
- Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Kendra I Melos
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Luise Angelini
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
41
|
Sessions GA, Copp ME, Liu JY, Sinkler MA, D'Costa S, Diekman BO. Controlled induction and targeted elimination of p16 INK4a-expressing chondrocytes in cartilage explant culture. FASEB J 2019; 33:12364-12373. [PMID: 31408372 DOI: 10.1096/fj.201900815rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cellular senescence is a phenotypic state that contributes to age-related diseases through the secretion of matrix-degrading and inflammatory molecules. An emerging therapeutic strategy for osteoarthritis (OA) is to selectively eliminate senescent cells by initiating apoptosis. This study establishes a cartilage explant model of senescence induction and senolytic clearance using p16Ink4a expression as a biomarker of senescence. Growth-factor stimulation of explants increased the expression of p16Ink4a at both the mRNA and protein levels. Applying this culture system to cartilage from p16tdTom reporter mice (a knockin allele with tdTomato fluorescent protein regulated by the endogenous p16Ink4a promoter) demonstrated the emergence of a p16-high population that was quantified using flow cytometry for tdTomato. Cell sorting was used to separate chondrocytes based on tdTomato fluorescence and p16-high cells showed higher senescence-associated β-galactosidase activity and increased gene expression of the senescence-associated secretory phenotype as compared with p16-low cells. The potential for effective senolysis within the cartilage extracellular matrix was assessed using navitoclax (ABT-263). Navitoclax treatment reduced the percentage of p16-high cells from 17.9 to 6.1% (mean of 13 matched pairs; P < 0.001) and increased cleaved caspase-3 confirmed apoptotic activity. Together, these findings establish a physiologically relevant cartilage explant model for testing the induction and elimination of senescent chondrocytes, which will support investigations of senolytic therapy for OA.-Sessions, G. A., Copp, M. E., Liu, J.-Y., Sinkler, M. A., D'Costa, S., Diekman, B. O. Controlled induction and targeted elimination of p16INK4a-expressing chondrocytes in cartilage explant culture.
Collapse
Affiliation(s)
- Garrett A Sessions
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michaela E Copp
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina-North Carolina State University, Raleigh, North Carolina, USA
| | - Jie-Yu Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Margaret A Sinkler
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Susan D'Costa
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina-North Carolina State University, Raleigh, North Carolina, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
42
|
Maguire G. The Safe and Efficacious Use of Secretome From Fibroblasts and Adipose-derived (but not Bone Marrow-derived) Mesenchymal Stem Cells for Skin Therapeutics. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2019; 12:E57-E69. [PMID: 31531174 PMCID: PMC6715117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Stem cell-based products are rapidly emerging in the marketplace as topical skin care and wound care products. Confusion is prevalent among healthcare providers and end-users about these products. Adipose-derived stem cells, fibroblasts, platelets, and bone marrow-derived stem cells are the most common cells used for stem cell therapeutic development, medical procedures, and skin care products. In this review, the significant advantages of adipose-derived stem cells and fibroblasts in terms of safety and efficacy are highlighted and compared to relatively risky platelets and bone marrow stem cells.
Collapse
Affiliation(s)
- Greg Maguire
- Dr. Maguire is with NeoGenesis, Inc. in San Diego, California
| |
Collapse
|
43
|
Maguire G, Paler L, Green L, Mella R, Valcarcel M, Villace P. Rescue of degenerating neurons and cells by stem cell released molecules: using a physiological renormalization strategy. Physiol Rep 2019; 7:e14072. [PMID: 31050222 PMCID: PMC6497969 DOI: 10.14814/phy2.14072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 12/13/2022] Open
Abstract
Evidence suggests that adult stem cell types and progenitor cells act collectively in a given tissue to maintain and heal organs, such as muscle, through a release of a multitude of molecules packaged into exosomes from the different cell types. Using this principle for the development of bioinspired therapeutics that induces homeostatic renormalization, here we show that the collection of molecules released from four cell types, including mesenchymal stem cells, fibroblast, neural stem cells, and astrocytes, rescues degenerating neurons and cells. Specifically, oxidative stress induced in a human recombinant TDP-43- or FUS-tGFP U2OS cell line by exposure to sodium arsenite was shown to be significantly reduced by our collection of molecules using in vitro imaging of FUS and TDP-43 stress granules. Furthermore, we also show that the collective secretome rescues cortical neurons from glutamate toxicity as evidenced by increased neurite outgrowth, reduced LDH release, and reduced caspase 3/7 activity. These data are the first in a series supporting the development of stem cell-based exosome systems therapeutics that uses a physiological renormalization strategy to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences, Inc.San DiegoCalifornia
- Auditory Sound Waves, LLCSan DiegoCalifornia
| | - Lee Paler
- BioRegenerative Sciences, Inc.San DiegoCalifornia
- Auditory Sound Waves, LLCSan DiegoCalifornia
| | - Linda Green
- BioRegenerative Sciences, Inc.San DiegoCalifornia
| | | | | | | |
Collapse
|
44
|
Knaus HA, Berglund S, Hackl H, Blackford AL, Zeidner JF, Montiel-Esparza R, Mukhopadhyay R, Vanura K, Blazar BR, Karp JE, Luznik L, Gojo I. Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight 2018; 3:120974. [PMID: 30385732 DOI: 10.1172/jci.insight.120974] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 09/19/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Our understanding of phenotypic and functional signatures of CD8+ T cell dysfunction in acute myeloid leukemia (AML) is limited. Deciphering these deranged T cell functional states and how they are impacted by induction chemotherapy is essential for incorporation of novel immune-based strategies to restore and maintain antileukemia immunity. METHODS We utilized high-dimensional immunophenotyping, gene expression, and functional studies to characterize peripheral blood and bone marrow CD8+ T cells in 72 AML patients at diagnosis and after induction chemotherapy. RESULTS Our data suggest that multiple aspects of deranged T cell function are operative in AML at diagnosis, with exhaustion and senescence being the dominant processes. Following treatment, the phenotypic and transcriptional profile of CD8+ T cells diverged between responders and nonresponders. Response to therapy correlated with upregulation of costimulatory, and downregulation of apoptotic and inhibitory, T cell signaling pathways, indicative of restoration of T cell function. In functional studies, AML blasts directly altered CD8+ T cell viability, expansion, co-signaling and senescence marker expression. This CD8+ T cell dysfunction was in part reversible upon PD-1 blockade or OX40 costimulation in vitro. CONCLUSION Our findings highlight the uniqueness of AML in sculpting CD8+ T cell responses and the plasticity of their signatures upon chemotherapy response, providing a compelling rationale for integration of novel immunotherapies to augment antileukemia immunity. FUNDING This work was supported by the Leukemia & Lymphoma Society grant no. 6449-13; NIH grants UM1-CA186691 and R01-HL110907-01; the American Society for Blood and Marrow Transplantation New Investigator Award/Gabrielle's Angel Foundation; the Vienna Fund for Innovative Cancer Research; and by fellowships from the Wenner-Gren Foundation and the Swedish Society for Medical Research.
Collapse
Affiliation(s)
- Hanna A Knaus
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sofia Berglund
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hubert Hackl
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Amanda L Blackford
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joshua F Zeidner
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Raúl Montiel-Esparza
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rupkatha Mukhopadhyay
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Katrina Vanura
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Judith E Karp
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Leo Luznik
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ivana Gojo
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Knaus HA, Berglund S, Hackl H, Blackford AL, Zeidner JF, Montiel-Esparza R, Mukhopadhyay R, Vanura K, Blazar BR, Karp JE, Luznik L, Gojo I. Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight 2018. [PMID: 30385732 DOI: 10.1172/jci.insight.120974:e120974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Our understanding of phenotypic and functional signatures of CD8+ T cell dysfunction in acute myeloid leukemia (AML) is limited. Deciphering these deranged T cell functional states and how they are impacted by induction chemotherapy is essential for incorporation of novel immune-based strategies to restore and maintain antileukemia immunity. METHODS We utilized high-dimensional immunophenotyping, gene expression, and functional studies to characterize peripheral blood and bone marrow CD8+ T cells in 72 AML patients at diagnosis and after induction chemotherapy. RESULTS Our data suggest that multiple aspects of deranged T cell function are operative in AML at diagnosis, with exhaustion and senescence being the dominant processes. Following treatment, the phenotypic and transcriptional profile of CD8+ T cells diverged between responders and nonresponders. Response to therapy correlated with upregulation of costimulatory, and downregulation of apoptotic and inhibitory, T cell signaling pathways, indicative of restoration of T cell function. In functional studies, AML blasts directly altered CD8+ T cell viability, expansion, co-signaling and senescence marker expression. This CD8+ T cell dysfunction was in part reversible upon PD-1 blockade or OX40 costimulation in vitro. CONCLUSION Our findings highlight the uniqueness of AML in sculpting CD8+ T cell responses and the plasticity of their signatures upon chemotherapy response, providing a compelling rationale for integration of novel immunotherapies to augment antileukemia immunity. FUNDING This work was supported by the Leukemia & Lymphoma Society grant no. 6449-13; NIH grants UM1-CA186691 and R01-HL110907-01; the American Society for Blood and Marrow Transplantation New Investigator Award/Gabrielle's Angel Foundation; the Vienna Fund for Innovative Cancer Research; and by fellowships from the Wenner-Gren Foundation and the Swedish Society for Medical Research.
Collapse
Affiliation(s)
- Hanna A Knaus
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sofia Berglund
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hubert Hackl
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Amanda L Blackford
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joshua F Zeidner
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Raúl Montiel-Esparza
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rupkatha Mukhopadhyay
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Katrina Vanura
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Judith E Karp
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Leo Luznik
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ivana Gojo
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Relationship between Aging and Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2018; 24:1965-1970. [PMID: 30130587 DOI: 10.1016/j.bbmt.2018.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023]
Abstract
Hematopoietic cell transplantation (HCT) is increasingly utilized as a treatment for malignancies in the elderly population. At the same time, research has elucidated the impacts of HCT on bone marrow progenitor cells, one of which is accelerated aging. Clonal hematopoiesis has also been observed to occur in the aging population, both with and without HCT. The interplay between natural aging, clonal hemoatpoiesis, and the effects of HCT on the bone marrow, has not yet been addressed. Herein we explore this relationship, and its important clinical implications.
Collapse
|
47
|
Diekman BO, Sessions GA, Collins JA, Knecht AK, Strum SL, Mitin NK, Carlson CS, Loeser RF, Sharpless NE. Expression of p16 INK 4a is a biomarker of chondrocyte aging but does not cause osteoarthritis. Aging Cell 2018; 17:e12771. [PMID: 29744983 PMCID: PMC6052464 DOI: 10.1111/acel.12771] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2018] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence drives a functional decline of numerous tissues with aging by limiting regenerative proliferation and/or by producing pro‐inflammatory molecules known as the senescence‐associated secretory phenotype (SASP). The senescence biomarker p16INK4a is a potent inhibitor of the cell cycle but is not essential for SASP production. Thus, it is unclear whether p16INK4a identifies senescence in hyporeplicative cells such as articular chondrocytes and whether p16INK4a contributes to pathologic characteristics of cartilage aging. To address these questions, we examined the role of p16INK4a in murine and human models of chondrocyte aging. We observed that p16INK4amRNA expression was significantly upregulated with chronological aging in murine cartilage (~50‐fold from 4 to 18 months of age) and in primary human chondrocytes from 57 cadaveric donors (r2 = .27, p < .0001). Human chondrocytes exhibited substantial replicative potential in vitro that depended on the activity of cyclin‐dependent kinases 4 or 6 (CDK4/6), and proliferation was reduced in cells from older donors with increased p16INK4a expression. Moreover, increased chondrocyte p16INK4a expression correlated with several SASP transcripts. Despite the relationship between p16INK4a expression and these features of senescence, somatic inactivation of p16INK4a in chondrocytes of adult mice did not mitigate SASP expression and did not alter the rate of osteoarthritis (OA) with physiological aging or after destabilization of the medial meniscus. These results establish that p16INK4a expression is a biomarker of dysfunctional chondrocytes, but that the effects of chondrocyte senescence on OA are more likely driven by production of SASP molecules than by loss of chondrocyte replicative function.
Collapse
Affiliation(s)
- Brian O. Diekman
- Lineberger Comprehensive Cancer Center; University of North Carolina School of Medicine; Chapel Hill North Carolina
- Thurston Arthritis Research Center; University of North Carolina School of Medicine; Chapel Hill North Carolina
- Department of Biomedical Engineering; University of North Carolina, Chapel Hill, NC; North Carolina State University; Raleigh North Carolina
| | - Garrett A. Sessions
- Thurston Arthritis Research Center; University of North Carolina School of Medicine; Chapel Hill North Carolina
| | - John A. Collins
- Thurston Arthritis Research Center; University of North Carolina School of Medicine; Chapel Hill North Carolina
| | - Anne K. Knecht
- HealthSpan Diagnostics LLC; Research Triangle Park North Carolina
| | - Susan L. Strum
- HealthSpan Diagnostics LLC; Research Triangle Park North Carolina
| | - Natalia K. Mitin
- HealthSpan Diagnostics LLC; Research Triangle Park North Carolina
| | - Cathy S. Carlson
- Department of Veterinary Clinical Sciences; University of Minnesota; St. Paul Minnesota
| | - Richard F. Loeser
- Thurston Arthritis Research Center; University of North Carolina School of Medicine; Chapel Hill North Carolina
- Division of Rheumatology, Allergy, and Immunology; University of North Carolina School of Medicine; Chapel Hill North Carolina
| | - Norman E. Sharpless
- Lineberger Comprehensive Cancer Center; University of North Carolina School of Medicine; Chapel Hill North Carolina
- Departments of Medicine and Genetics; University of North Carolina School of Medicine; Chapel Hill North Carolina
- The National Cancer Institute; Bethesda Maryland
| |
Collapse
|
48
|
Massey JC, Sutton IJ, Ma DDF, Moore JJ. Regenerating Immunotolerance in Multiple Sclerosis with Autologous Hematopoietic Stem Cell Transplant. Front Immunol 2018; 9:410. [PMID: 29593711 PMCID: PMC5857574 DOI: 10.3389/fimmu.2018.00410] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/14/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disorder of the central nervous system where evidence implicates an aberrant adaptive immune response in the accrual of neurological disability. The inflammatory phase of the disease responds to immunomodulation to varying degrees of efficacy; however, no therapy has been proven to arrest progression of disability. Recently, more intensive therapies, including immunoablation with autologous hematopoietic stem cell transplantation (AHSCT), have been offered as a treatment option to retard inflammatory disease, prior to patients becoming irreversibly disabled. Empirical clinical observations support the notion that the immune reconstitution (IR) that occurs following AHSCT is associated with a sustained therapeutic benefit; however, neither the pathogenesis of MS nor the mechanism by which AHSCT results in a therapeutic benefit has been clearly delineated. Although the antigenic target of the aberrant immune response in MS is not defined, accumulated data suggest that IR following AHSCT results in an immunotolerant state through deletion of pathogenic clones by a combination of direct ablation and induction of a lymphopenic state driving replicative senescence and clonal attrition. Restoration of immunoregulation is evidenced by changes in regulatory T cell populations following AHSCT and normalization of genetic signatures of immune homeostasis. Furthermore, some evidence exists that AHSCT may induce a rebooting of thymic function and regeneration of a diversified naïve T cell repertoire equipped to appropriately modulate the immune system in response to future antigenic challenge. In this review, we discuss the immunological mechanisms of IR therapies, focusing on AHSCT, as a means of recalibrating the dysfunctional immune response observed in MS.
Collapse
Affiliation(s)
- Jennifer C Massey
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Neurology, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Ian J Sutton
- Neurology, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - David D F Ma
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - John J Moore
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
49
|
Maguire G. Amyotrophic lateral sclerosis as a protein level, non-genomic disease: Therapy with S2RM exosome released molecules. World J Stem Cells 2017; 9:187-202. [PMID: 29312526 PMCID: PMC5745587 DOI: 10.4252/wjsc.v9.i11.187] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/10/2017] [Accepted: 09/04/2017] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease that leads to death. No effective treatments are currently available. Based on data from epidemiological, etiological, laboratory, and clinical studies, I offer a new way of thinking about ALS and its treatment. This paper describes a host of extrinsic factors, including the exposome, that disrupt the extracellular matrix and protein function such that a spreading, prion-like disease leads to neurodegeneration in the motor tracts. A treatment regimen is described using the stem cell released molecules from a number of types of adult stem cells to provide tissue dependent molecules that restore homeostasis, including proteostasis, in the ALS patient. Because stem cells themselves as a therapeutic are cumbersome and expensive, and when implanted in a host cause aging of the host tissue and often fail to engraft or remain viable, only the S2RM molecules are used. Rebuilding of the extracellular matrix and repair of the dysfunctional proteins in the ALS patient ensues.
Collapse
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences, Inc., La Jolla, CA 92037, United States
| |
Collapse
|
50
|
Frailty: the missing piece of the pre- hematopoietic cell transplantation assessment? Bone Marrow Transplant 2017; 53:3-10. [PMID: 29084201 DOI: 10.1038/bmt.2017.192] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/22/2017] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) represents a curative option for those afflicted with numerous hematologic malignancies and bone marrow failure syndromes. Advances and refinement of the HSCT process have resulted in increasing number of transplants performed on older patients in the recent years. Pre-transplant assessments (PTA) function to risk stratify patients prior to undergoing HSCT in an effort to predict those at higher risk of treatment-related toxicity, to inform risk/benefit assessments and to aid clinical decision making. Traditionally used risk stratification parameters such as chronologic age, comorbidity and performance status may not fully capture physical function, physiologic fitness, highlighting a need for improvement in PTA. Incorporation of frailty measurements in pre-HSCT assessments, particularly in elderly transplant candidates, may result in improving predictive ability of existing tools such as the Hematopoietic Cell Transplantation Comorbidity Index and Karnofsky performance status. Here, we review existing pre-HSCT assessment tools, measures of frailty that may aid in risk stratification for patients undergoing HSCT and directions for future research using frailty in the pre-HSCT setting.
Collapse
|