Is less always more? Emerging treatment concepts in geriatric hemato-oncology

Authors

  • Nina Rosa Neuendorff Department for Hematology and Stem Cell Transplantation, University Hospital Essen, Hufelandstrasse 55, D-45147 Essen, Germany.
  • Hans Christian Reinhardt Department for Hematology and Stem Cell Transplantation, University Hospital Essen, Hufelandstrasse 55, D-45147 Essen, Germany.
  • Konstantinos Christofyllakis Department for Oncology, Hematology, Clinical Immunology and Rheumatology, Saarland University Medical School, Homburg/Saar, Germany

DOI:

https://doi.org/10.37155/2972-4759-2023-01-01-2

Keywords:

Cancer, Older adults, Comprehensive geriatric assessment, Chemotherapy, Sarcopenia, Undertreatment, Overtreatment

Abstract

The outcomes of older adults with cancer are still dismal despite some progress within the last years. This is mainly due to comorbidities, overall frailty, and differences in disease biology. The better understanding of tumor biology and immunology has enabled the use of targeted therapies and immunotherapies that are potentially better tolerated than traditional chemotherapies. Several randomized trials were recently published that demonstrated a benefit of integrated onco-geriatric care including geriatric interventions on treatment-related toxicities and quality of life during cancer treatment of older adults. Furthermore, other pivotal trials adapted treatment intensities based on frailty. Despite these efforts, older and frail adults are still underrepresented in clinical trials. This leads to a major lack of an evidence-based standard of care in geriatric oncology.

In this narrative review, we discuss pivotal trials, practical implications, under- and overtreatment, altered pharmacokinetics at older age, and future perspectives for geriatric oncology.

References

Goede V, Neuendorff N R, Schulz R J, et al. Frailty assessment in the care of older people with haematological malignancies[J]. The Lancet Healthy Longevity, 2021, 2(11): e736-e745.

https://doi.org/10.1016/S2666-7568(21)00184-7

Van Herck Y, Feyaerts A, Alibhai S, et al. Is cancer biology different in older patients?[J]. The Lancet Healthy Longevity, 2021, 2(10): e663-e677.

https://doi.org/10.1016/S2666-7568(21)00179-3

Soto-Perez-de-Celis E, Li D, Yuan Y, et al. Functional versus chronological age: geriatric assessments to guide decision making in older patients with cancer[J]. The Lancet Oncology, 2018, 19(6): e305-e316.

https://doi.org/10.1016/S1470-2045(18)30348-6

Mohile S G, Mohamed M R, Xu H, et al. Evaluation of geriatric assessment and management on the toxic effects of cancer treatment (GAP70+): a cluster-randomised study[J]. The Lancet, 2021, 398(10314): 1894-1904.

https://doi.org/10.1016/S0140-6736(21)01789-X

Li D, Sun C L, Kim H, et al. Geriatric assessment–driven intervention (GAIN) on chemotherapy-related toxic effects in older adults with cancer: a randomized clinical trial[J]. JAMA oncology, 2021, 7(11): e214158-e214158.

https://doi.org/10.1001/jamaoncol.2021.4158

Soo W K, King M T, Pope A, et al. Integrated Geriatric Assessment and Treatment Effectiveness (INTEGERATE) in older people with cancer starting systemic anticancer treatment in Australia: a multicentre, open-label, randomised controlled trial[J]. The Lancet Healthy Longevity, 2022, 3(9): e617-e627.

https://doi.org/10.1016/S2666-7568(22)00169-6

Mohile S G, Dale W, Somerfield M R, et al. Practical assessment and management of vulnerabilities in older patients receiving chemotherapy: ASCO guideline for geriatric oncology[J]. Journal of Clinical Oncology, 2018, 36(22): 2326-2347.

https://doi.org/10.1200/JCO.2018.78.8687

Dotan E, Walter L C, Browner I S, et al. NCCN guidelines® insights: older adult oncology, version 1.2021: featured updates to the NCCN guidelines[J]. Journal of the National Comprehensive Cancer Network, 2021, 19(9): 1006-1019.

https://doi.org/10.6004/jnccn.2021.0043

Decoster L, Van Puyvelde K, Mohile S, et al. Screening tools for multidimensional health problems warranting a geriatric assessment in older cancer patients: an update on SIOG recommendations[J]. Annals of Oncology, 2015, 26(2): 288-300.

https://doi.org/10.1093/annonc/mdu210

Uranga C, Chien L C, Liposits G. Geriatric screening in older adults with cancer–A Young International Society of Geriatric Oncology and Nursing & Allied Health Interest Group initiative[J]. Journal of Geriatric Oncology, 2022, 13(3): 374-377.

https://doi.org/10.1016/j.jgo.2021.09.003

Bellera C A, Rainfray M, Mathoulin-Pélissier S, et al. Screening older cancer patients: first evaluation of the G-8 geriatric screening tool[J]. Annals of Oncology, 2012, 23(8): 2166-2172.

https://doi.org/10.1093/annonc/mdr587

Kenis C, Decoster L, Van Puyvelde K, et al. Performance of two geriatric screening tools in older patients with cancer[J]. Journal of Clinical Oncology, 2014, 32(1): 19-26.

https://doi.org/10.1200/JCO.2013.51.1345

Oiwa K, Fujita K, Lee S, et al. Utility of the geriatric 8 for the prediction of therapy‐related toxicity in older adults with diffuse large B‐cell lymphoma[J]. The Oncologist, 2021, 26(3): 215-223.

https://doi.org/10.1002/onco.13641

Lund C M, Vistisen K K, Olsen A P, et al. The effect of geriatric intervention in frail older patients receiving chemotherapy for colorectal cancer: a randomised trial (GERICO)[J]. British Journal of Cancer, 2021, 124(12): 1949-1958.

https://doi.org/10.1038/s41416-021-01367-0

Saliba D, Elliott M, Rubenstein L Z, et al. The Vulnerable Elders Survey: a tool for identifying vulnerable older people in the community[J]. Journal of the American Geriatrics Society, 2001, 49(12): 1691-1699.

https://doi.org/10.1046/j.1532-5415.2001.49281.x

Pottel L, Boterberg T, Pottel H, et al. Determination of an adequate screening tool for identification of vulnerable elderly head and neck cancer patients treated with radio (chemo) therapy[J]. Journal of geriatric oncology, 2012, 3(1): 24-32.

https://doi.org/10.1016/j.jgo.2011.11.006

Hurria A, Togawa K, Mohile S G, et al. Predicting chemotherapy toxicity in older adults with cancer: a prospective multicenter study[J]. Journal of clinical oncology, 2011, 29(25): 3457.

https://doi.org/10.1200/JCO.2011.34.7625

Hurria A, Mohile S, Gajra A, et al. Validation of a prediction tool for chemotherapy toxicity in older adults with cancer[J]. Journal of Clinical Oncology, 2016, 34(20): 2366.

https://doi.org/10.1200/JCO.2015.65.4327

Extermann M, Boler I, Reich R R, et al. Predicting the risk of chemotherapy toxicity in older patients: The Chemotherapy Risk Assessment Scale for High‐Age Patients (CRASH) score[J]. Cancer, 2012, 118(13): 3377-3386.

https://doi.org/10.1002/cncr.26646

Battisti N M L, Arora S P. An overview of chemotherapy toxicity prediction tools in older adults with cancer: A young international society of geriatric oncology and nursing and allied health initiative[J]. Journal of Geriatric Oncology, 2022, 13(4): 521-525.

https://doi.org/10.1016/j.jgo.2021.12.006

Kuon J, Hommertgen A, Krisam J, et al. Durvalumab in frail and elderly patients with stage four non-small cell lung cancer: Study protocol of the randomized phase II DURATION trial[J]. Trials, 2020, 21: 352.

https://doi.org/10.1186/s13063-020-04280-8

Ritchie E K, Klepin H D, Storrick E, et al. Geriatric assessment for older adults receiving less-intensive therapy for acute myeloid leukemia: report of CALGB 361101[J]. Blood Advances, 2022, 6(12): 3812-3820.

https://doi.org/10.1182/bloodadvances.2021006872

Klepin H D, Ritchie E, Major-Elechi B, et al. Geriatric assessment among older adults receiving intensive therapy for acute myeloid leukemia: report of CALGB 361006 (Alliance)[J]. Journal of geriatric oncology, 2020, 11(1): 107-113.

https://doi.org/10.1016/j.jgo.2019.10.002

DuMontier C, Uno H, Hshieh T, et al. Randomized controlled trial of geriatric consultation versus standard care in older adults with hematologic malignancies[J]. Haematologica, 2022, 107(5): 1172-1180.

https://doi.org/10.3324/haematol.2021.278802

Puts M, Alqurini N, Strohschein F, et al. Impact of Geriatric Assessment and Management on Quality of Life, Unplanned Hospitalizations, Toxicity, and Survival for Older Adults With Cancer: The Randomized 5C Trial[J]. Journal of Clinical Oncology, 2022: JCO. 22.01007.

https://doi.org/10.1200/JCO.22.01007

Cruz-Jentoft A J, Bahat G, Bauer J, et al. Sarcopenia: revised European consensus on definition and diagnosis[J]. Age and ageing, 2019, 48(1): 16-31.

https://doi.org/10.1093/ageing/afy169

Anjanappa M, Corden M, Green A, et al. Sarcopenia in cancer: Risking more than muscle loss[J]. Technical innovations & patient support in radiation oncology, 2020, 16: 50-57.

https://doi.org/10.1016/j.tipsro.2020.10.001

Malmstrom T K, Miller D K, Simonsick E M, et al. SARC‐F: a symptom score to predict persons with sarcopenia at risk for poor functional outcomes[J]. Journal of cachexia, sarcopenia and muscle, 2016, 7(1): 28-36.

https://doi.org/10.1002/jcsm.12048

Maurus J, Terzer T, Benner A, et al. Validation of a proxy‐reported SARC‐F questionnaire for current and retrospective screening of sarcopenia‐related functional impairments[J]. Journal of Cachexia, Sarcopenia and Muscle, 2022, 13(1): 264-275.

https://doi.org/10.1002/jcsm.12871

Zhao H, Cheng R, Song G, et al. The Effect of Resistance Training on the Rehabilitation of Elderly Patients with Sarcopenia: A Meta-Analysis[J]. International Journal of Environmental Research and Public Health, 2022, 19(23): 15491.

https://doi.org/10.3390/ijerph192315491

Bauer J, Morley J E, Schols A M W J, et al. Sarcopenia: a time for action. An SCWD position paper[J]. Journal of cachexia, sarcopenia and muscle, 2019, 10(5): 956-961.

https://doi.org/10.1002/jcsm.12483

Tan B H L, Brammer K, Randhawa N, et al. Sarcopenia is associated with toxicity in patients undergoing neo-adjuvant chemotherapy for oesophago-gastric cancer[J]. European Journal of Surgical Oncology (EJSO), 2015, 41(3): 333-338.

https://doi.org/10.1016/j.ejso.2014.11.040

Jung H W, Kim J W, Kim J Y, et al. Effect of muscle mass on toxicity and survival in patients with colon cancer undergoing adjuvant chemotherapy[J]. Supportive care in cancer, 2015, 23: 687-694.

https://doi.org/10.1007/s00520-014-2418-6

Prado C M M, Baracos V E, McCargar L J, et al. Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment[J]. Clinical cancer research, 2009, 15(8): 2920-2926.

https://doi.org/10.1158/1078-0432.CCR-08-2242

Matsunaga T, Saito H, Miyauchi W, et al. Impact of skeletal muscle mass in patients with unresectable gastric cancer who received palliative first-line chemotherapy based on 5-fluorouracil[J]. BMC cancer, 2021, 21(1): 1219.

https://doi.org/10.1186/s12885-021-08953-8

Halliday L J, Boshier P R, Doganay E, et al. The effects of prehabilitation on body composition in patients undergoing multimodal therapy for esophageal cancer[J]. Diseases of the Esophagus, 2023, 36(2): doac046.

https://doi.org/10.1093/dote/doac046

Roche M, Ravot C, Malapert A, et al. Feasibility of a prehabilitation programme dedicated to older patients with cancer before complex medical–surgical procedures: The PROADAPT pilot study protocol[J]. BMJ open, 2021, 11(4): e042960.

http://dx.doi.org/10.1136/bmjopen-2020-042960

Dolin T G, Mikkelsen M, Jakobsen H L, et al. Geriatric assessment and intervention in older vulnerable patients undergoing surgery for colorectal cancer: a protocol for a randomised controlled trial (GEPOC trial)[J]. BMC geriatrics, 2021, 21(1): 1-10.

https://doi.org/10.1186/s12877-021-02045-9

Collins K H, Herzog W, MacDonald G Z, et al. Obesity, metabolic syndrome, and musculoskeletal disease: common inflammatory pathways suggest a central role for loss of muscle integrity[J]. Frontiers in physiology, 2018, 9: 112.

https://doi.org/10.3389/fphys.2018.00112

Crochow L B, Baraldi C, Noe D. Is dose normalization to weight or body surface area useful in adults?[J]. JNCI: Journal of the National Cancer Institute, 1990, 82(4): 323-325.

https://doi.org/10.1093/jnci/82.4.323

Baker S D, Verweij J, Rowinsky E K, et al. Role of body surface area in dosing of investigational anticancer agents in adults, 1991–2001[J]. Journal of the National Cancer Institute, 2002, 94(24): 1883-1888.

https://doi.org/10.1093/jnci/94.24.1883

Williams G R, Outlaw D, Harvey R D, et al. Chemotherapy dosing in older adults with cancer: One size does NOT fit all[J]. Journal of Geriatric Oncology, 2023, 14(1).

https://doi.org/10.1016/j.jgo.2022.08.012

Hertz D L, Chen L, Henry N L, et al. Muscle mass affects paclitaxel systemic exposure and may inform personalized paclitaxel dosing[J]. British journal of clinical pharmacology, 2022, 88(7): 3222-3229.

https://doi.org/10.1111/bcp.15244

Chargi N, Molenaar-Kuijsten L, Huiskamp L F J, et al. The association of cisplatin pharmacokinetics and skeletal muscle mass in patients with head and neck cancer: the prospective PLATISMA study[J]. European Journal of Cancer, 2022, 160: 92-99.

https://doi.org/10.1016/j.ejca.2021.10.010

Drami I, Pring E T, Gould L, et al. Body composition and dose-limiting toxicity in colorectal cancer chemotherapy treatment; a systematic review of the literature. Could muscle mass be the new body surface area in chemotherapy dosing?[J]. Clinical Oncology, 2021, 33(12): e540-e552.

https://doi.org/10.1016/j.clon.2021.05.011

Hall P S, Swinson D, Cairns D A, et al. Efficacy of reduced-intensity chemotherapy with oxaliplatin and capecitabine on quality of life and cancer control among older and frail patients with advanced gastroesophageal cancer: the GO2 phase 3 randomized clinical trial[J]. JAMA oncology, 2021, 7(6): 869-877.

https://doi.org/10.1001/jamaoncol.2021.0848

Lund C M, Vistisen K K, Dehlendorff C, et al. The effect of geriatric intervention in frail elderly patients receiving chemotherapy for colorectal cancer: a randomized trial (GERICO)[J]. BMC cancer, 2017, 17(1): 1-9.

https://doi.org/10.1186/s12885-017-3445-8

Grant S J, Mian H S, Giri S, et al. Transplant-ineligible newly diagnosed multiple myeloma: Current and future approaches to clinical care: A Young International Society of Geriatric Oncology Review Paper[J]. Journal of geriatric oncology, 2021, 12(4): 499-507.

https://doi.org/10.1016/j.jgo.2020.12.001

Klepin H D, Estey E, Kadia T. More versus less therapy for older adults with acute myeloid leukemia: new perspectives on an old debate[J]. American Society of Clinical Oncology Educational Book, 2019, 39: 421-432.

https://doi.org/10.1200/EDBK_239097

Klepin H D, Neuendorff N R, Larson R A, et al. Treatment of acute promyelocytic leukemia in older patients: recommendations of an International Society of Geriatric Oncology (SIOG) task force[J]. Journal of Geriatric Oncology, 2020, 11(8): 1199-1209.

https://doi.org/10.1016/j.jgo.2020.03.019

Burnett A K, Milligan D, Prentice A G, et al. A comparison of low‐dose cytarabine and hydroxyurea with or without all‐trans retinoic acid for acute myeloid leukemia and high‐risk myelodysplastic syndrome in patients not considered fit for intensive treatment[J]. Cancer: Interdisciplinary International Journal of the American Cancer Society, 2007, 109(6): 1114-1124.

https://doi.org/10.1002/cncr.22496

Neuendorff N R, Gagelmann N, Singhal S, et al. Hypomethylating agent-based therapies in older adults with acute myeloid leukemia–A joint review by the Young International Society of Geriatric Oncology and European Society for Blood and Marrow Transplantation Trainee Committee[J]. Journal of Geriatric Oncology, 2022.

https://doi.org/10.1016/j.jgo.2022.11.005

Récher C, Röllig C, Bérard E, et al. Long-term survival after intensive chemotherapy or hypomethylating agents in AML patients aged 70 years and older: a large patient data set study from European registries[J]. Leukemia, 2022. 36(4): p. 913-922.

https://doi.org/10.1038/s41375-021-01425-9

Richardson, Daniel R., et al. "Time at home among older adults with acute myeloid leukemia based on treatment intensity: A SEER-Medicare analysis." (2022): 6586-6586.

https://doi.org/10.1200/JCO.2022.40.16_suppl.6586

DiNardo C D, Jonas B A, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia[J]. New England Journal of Medicine, 2020, 383(7): 617-629.

https://doi.org/10.1056/NEJMoa2012971

Pratz K W, Panayiotidis P, Recher C, et al. Venetoclax combinations delay the time to deterioration of HRQoL in unfit patients with acute myeloid leukemia[J]. Blood cancer journal, 2022, 12(4): 71.

https://doi.org/10.1038/s41408-022-00668-8

Montesinos P, Recher C, Vives S, et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia[J]. New England Journal of Medicine, 2022, 386(16): 1519-1531.

https://doi.org/10.1056/NEJMoa2117344

Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma[J]. New England Journal of Medicine, 2019, 380(22): 2104-2115.

https://doi.org/10.1056/NEJMoa1817249

Facon T, Cook G, Usmani S Z, et al. Daratumumab plus lenalidomide and dexamethasone in transplant-ineligible newly diagnosed multiple myeloma: frailty subgroup analysis of MAIA[J]. Leukemia, 2022, 36(4): 1066-1077.

https://doi.org/10.1038/s41375-021-01488-8

Perrot A, Facon T, Plesner T, et al. Health-related quality of life in transplant-ineligible patients with newly diagnosed multiple myeloma: findings from the phase III MAIA trial[J]. Journal of Clinical Oncology, 2021, 39(3): 227.

https://doi.org/10.1200/JCO.20.01370

Li P, Yang X, Feng Y, et al. The impact of immunosenescence on the efficacy of immune checkpoint inhibitors in melanoma patients: a meta-analysis[J]. OncoTargets and therapy, 2018: 7521-7527.

https://doi.org/10.2147/OTT.S165368

Kanesvaran R, Cordoba R, Maggiore R. Immunotherapy in older adults with advanced cancers: implications for clinical decision-making and future research[J]. American Society of Clinical Oncology Educational Book, 2018, 38: 400-414.

https://doi.org/10.1200/EDBK_201435

Tawbi H A, Schadendorf D, Lipson E J, et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma[J]. New England Journal of Medicine, 2022, 386(1): 24-34.

https://doi.org/10.1056/NEJMoa2109970

Wei J, Yang Y, Wang G, et al. Current landscape and future directions of bispecific antibodies in cancer immunotherapy[J]. Frontiers in Immunology, 2022, 13: 6573.

https://doi.org/10.3389/fimmu.2022.1035276

Joubert, N., et al., Antibody–Drug Conjugates: The Last Decade. Pharmaceuticals, 2020. 13(9): p. 245.

https://doi.org/10.3390/ph13090245

Fitzgerald L, Kittai A, Nastoupil L J, et al. Real-world outcomes of elderly patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) treated with chimeric antigen receptor T-cell (CAR-T) therapy[J]. Journal of Clinical Oncology, 2020. 38: p. 8039-8039.

https://doi.org/10.1200/JCO.2020.38.15_suppl.8039

Lin R J, Lobaugh S M, Pennisi M, et al. Impact and safety of chimeric antigen receptor T-cell therapy in older, vulnerable patients with relapsed/refractory large B-cell lymphoma[J]. Haematologica, 2021, 106(1): 255-258.

https://doi.org/10.3324/haematol.2019.243246

Berdeja J G, Madduri D, Usmani S Z, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study[J]. The Lancet, 2021, 398(10297): 314-324.

https://doi.org/10.1016/S0140-6736(21)00933-8

Munshi N C, Anderson Jr L D, Shah N, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma[J]. New England Journal of Medicine, 2021, 384(8): 705-716.

https://doi.org/10.1056/NEJMoa2024850

Berdeja J G, Raje N S, Siegel D S, et al. Efficacy and safety of idecabtagene vicleucel (ide-cel, bb2121) in elderly patients with relapsed and refractory multiple myeloma: karMMa subgroup analysis[J]. Blood, 2020, 136: 16-17.

https://doi.org/10.1182/blood-2020-134322

Shouse G, Danilov A V, Artz A. CAR T-cell therapy in the older person: indications and risks[J]. Current Oncology Reports, 2022, 24(9): 1189-1199.

https://doi.org/10.1007/s11912-022-01272-6

Giri S, Bal S, Godby K N, et al. Real-world applicability of commercial chimeric antigen receptor T-cell therapy among older adults with relapsed and/or refractory multiple myeloma[J]. American journal of hematology, 2022, 97(4): E153-E155.

https://doi.org/10.1002/ajh.26472

Kaiser M, Semeraro M D, Herrmann M, et al. Immune aging and immunotherapy in cancer[J]. International Journal of Molecular Sciences, 2021, 22(13): 7016.

https://doi.org/10.3390/ijms22137016

Wang C, Wang H, Wang L. Biomarkers for predicting the efficacy of immune checkpoint inhibitors[J]. Journal of Cancer, 2022, 13(2): 481-495.

https://doi.org/10.7150/jca.65012

Moreira A, Gross S, Kirchberger M C, et al. Senescence markers: Predictive for response to checkpoint inhibitors[J]. International journal of cancer, 2019, 144(5): 1147-1150.

https://doi.org/10.1002/ijc.31763

Ferrara R, Naigeon M, Auclin E, et al. Circulating T-cell Immunosenescence in Patients with Advanced Non–small Cell Lung Cancer Treated with Single-agent PD-1/PD-L1 Inhibitors or Platinum-based ChemotherapyImmunosenescence in NSCLC[J]. Clinical Cancer Research, 2021, 27(2): 492-503.

https://doi.org/10.1158/1078-0432.CCR-20-1420

Steensma D P, Bejar R, Jaiswal S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes[J]. Blood, The Journal of the American Society of Hematology, 2015, 126(1): 9-16.

https://doi.org/10.1182/blood-2015-03-631747

Neuendorff N R, Frenzel L P, Leuschner F, et al. Integrating clonal haematopoiesis into geriatric oncology: The ARCH between aging, cardiovascular disease and malignancy[J]. Journal of Geriatric Oncology, 2021, 12(3): 479-482.

https://doi.org/10.1016/j.jgo.2020.09.006

BrintzenhofeSzoc K, Krok-Schoen J L, Canin B, et al. The underreporting of phase III chemo-therapeutic clinical trial data of older patients with cancer: A systematic review[J]. Journal of geriatric oncology, 2020, 11(3): 369-379.

https://doi.org/10.1016/j.jgo.2019.12.007

Hopkins J O, Braun-Inglis C, Guidice S, et al. Enrolling Older Adults Onto National Cancer Institute–Funded Clinical Trials in Community Oncology Clinics: Barriers and Solutions[J]. JNCI Monographs, 2022, 2022(60): 117-124.

https://doi.org/10.1093/jncimonographs/lgac019

Le-Rademacher J, Mohile S, Unger J, et al. Trial Design Considerations to Increase Older Adult Accrual to National Cancer Institute Clinical Trials[J]. JNCI Monographs, 2022, 2022(60): 135-141.

https://doi.org/10.1093/jncimonographs/lgac023

Richardson, D.R. and K.P. Loh, Improving personalized treatment decision-making for older adults with cancer: The necessity of eliciting patient preferences. J Geriatr Oncol, 2022. 13(1): p. 1-3.

https://doi.org/10.1016/j.jgo.2021.06.001

Downloads

Published

2023-05-22