Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.067
Filter
1.
Cancer Discov ; 14(6): 900-902, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38826099

ABSTRACT

SUMMARY: Given the rarity of cancer in childhood, it should be even more uncommon for pediatric cancer survivors to develop a second, independent malignancy, yet they incur a greatly elevated risk after initial remission. In this issue of Cancer Discovery, Sánchez-Guixé and colleagues unpick the origins of second tumours in four children, and the potential role platinum-based chemotherapy may play in subsequent tumorigenesis. See related article by Sánchez-Guixé et al., p. 953 (8).


Subject(s)
Neoplasms, Second Primary , Humans , Neoplasms, Second Primary/etiology , Neoplasms, Second Primary/epidemiology , Child , Cancer Survivors , Neoplasms/drug therapy
2.
Clin Respir J ; 18(5): e13760, 2024 May.
Article in English | MEDLINE | ID: mdl-38725324

ABSTRACT

OBJECTIVE: Radiation therapy (RT) may increase the risk of second cancer. This study aimed to determine the association between exposure to radiotherapy for the treatment of thoracic cancer (TC) and subsequent secondary lung cancer (SLC). MATERIALS AND METHODS: The Surveillance, Epidemiology, and End Results (SEER) database (from 1975 to 2015) was queried for TC. Univariate Cox regression analyses and multiple primary standardized incidence ratios (SIRs) were used to assess the risk of SLC. Subgroup analyses of patients stratified by latency time since TC diagnosis, age at TC diagnosis, and calendar year of TC diagnosis stage were also performed. Overall survival and SLC-related death were compared among the RT and no radiation therapy (NRT) groups by using Kaplan-Meier analysis and competitive risk analysis. RESULTS: In a total of 329 129 observations, 147 847 of whom had been treated with RT. And 6799 patients developed SLC. Receiving radiotherapy was related to a higher risk of developing SLC for TC patients (adjusted HR, 1.25; 95% CI, 1.19-1.32; P < 0.001). The cumulative incidence of developing SLC in TC patients with RT (3.8%) was higher than the cumulative incidence (2.9%) in TC patients with NRT(P). The incidence risk of SLC in TC patients who received radiotherapy was significantly higher than the US general population (SIR, 1.19; 95% CI, 1.14-1.23; P < 0.050). CONCLUSIONS: Radiotherapy for TC was associated with higher risks of developing SLC compared with patients unexposed to radiotherapy.


Subject(s)
Lung Neoplasms , Neoplasms, Second Primary , SEER Program , Thoracic Neoplasms , Humans , Male , Female , Lung Neoplasms/radiotherapy , Lung Neoplasms/epidemiology , Middle Aged , Aged , Incidence , Prognosis , Thoracic Neoplasms/radiotherapy , Thoracic Neoplasms/epidemiology , Neoplasms, Second Primary/epidemiology , Neoplasms, Second Primary/etiology , Retrospective Studies , Risk Factors , United States/epidemiology , Radiotherapy/adverse effects , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Risk Assessment/methods , Adult
5.
JNCI Cancer Spectr ; 8(3)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38718185

ABSTRACT

Type II diabetes is associated with cancer risk in the general population but has not been well studied as a risk factor for subsequent malignancies among cancer survivors. We investigated the association between diabetes and subsequent cancer risk among older (66-84 years), 1-year breast cancer survivors within the linked Surveillance Epidemiology and End Results (SEER)-Medicare database using Cox regression analyses to quantify hazard ratios (HR) and corresponding 95% confidence intervals (95% CI). Among 133 324 women, 29.3% were diagnosed with diabetes before or concurrent with their breast cancer diagnosis, and 10 452 women developed subsequent malignancies over a median follow-up of 4.3 years. Diabetes was statistically significantly associated with liver (HR = 2.35, 95% CI = 1.48 to 3.74), brain (HR = 1.94, 95% CI = 1.26 to 2.96), and thyroid cancer risks (HR = 1.38, 95% CI = 1.01 to 1.89). Future studies are needed to better understand the spectrum of subsequent cancers associated with diabetes and the role of diabetes medications in modifying subsequent cancer risk, alone or in combination with cancer treatments.


Subject(s)
Breast Neoplasms , Cancer Survivors , Diabetes Mellitus, Type 2 , Proportional Hazards Models , SEER Program , Humans , Female , Aged , Breast Neoplasms/epidemiology , Breast Neoplasms/etiology , Aged, 80 and over , Cancer Survivors/statistics & numerical data , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , United States/epidemiology , Risk Factors , Liver Neoplasms/epidemiology , Liver Neoplasms/etiology , Thyroid Neoplasms/epidemiology , Thyroid Neoplasms/etiology , Brain Neoplasms/epidemiology , Brain Neoplasms/etiology , Medicare/statistics & numerical data , Neoplasms, Second Primary/epidemiology , Neoplasms, Second Primary/etiology
6.
J Pak Med Assoc ; 74(5): 984-986, 2024 May.
Article in English | MEDLINE | ID: mdl-38783452

ABSTRACT

Acute promyelocytic leukaemia (APL) is a form of acute myelogenous leukaemia. APL is characterised by anaemia due to suppression of normal haematopoiesis and infection. Haematopoietic stem cell transplantation (HSCT) is current option for the treatment of haematopoietic malignancies and is proving to be successful. Although HSCT has been effective for the treatment of haematopoietic malignant tumours, chronic graft-versushost disease (GVHD) but secondary cancers can occur, which is a serious complication and frequently involves the oral cavity and skin. Here, we report the case of tongue cancer occurring 17 years after transplantation in a patient who developed GVHD after haematopoietic stem cell transplantation and APL remission. To the best of our knowledge, this is the first report of secondary oral cancer after HSCT with APL as the primary disease.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Promyelocytic, Acute , Tongue Neoplasms , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Leukemia, Promyelocytic, Acute/therapy , Tongue Neoplasms/surgery , Tongue Neoplasms/therapy , Male , Graft vs Host Disease/etiology , Middle Aged , Adult , Neoplasms, Second Primary/etiology
7.
Front Immunol ; 15: 1342845, 2024.
Article in English | MEDLINE | ID: mdl-38571955

ABSTRACT

Introduction: Over the past decade, immune checkpoint inhibitors such as antibodies against cytotoxicity T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) have become an important armamentarium against a broad spectrum of malignancies. However, these specific inhibitors can cause adverse autoimmune reactions by impairing self-tolerance. Hematologic side effects of immune checkpoint inhibitors, including autoimmune hemolytic anemia (AIHA), are rare but can be life-threatening. Case report: Herein, we report two patients on immune checkpoint inhibitors for metastatic melanoma who developed AIHA with symptoms of dyspnea and fatigue. In the first patient, symptoms alleviated after discontinuation of combined anti CTLA-4 and anti-PD-1 therapy, initiation of corticosteroids and application of a single red blood cell transfusion. Due to subsequent progress of melanoma, combinational anti-PD-1 and tyrosine kinase inhibitor therapy was initiated based on multidisciplinary tumor board decision. After two months, she again developed the described hematological and clinical signs of AIHA leading to cessation of anti-PD-1 therapy and initiation of corticosteroids, which again resulted in an alleviation of her symptoms. Due to further progression, the patient received dacarbazine for several months before she decided to stop any therapy other than palliative supportive care. In the second patient, discontinuation of anti-PD-1 therapy and initiation of corticosteroids entailed a complete alleviation of his symptoms. After refusing chemotherapy due to subsequent melanoma progression, he received radiotherapy of bone metastases and is currently enrolled in a clinical trial. The patient did not develop AIHA ever since. Conclusion: Hematologic immune-related adverse events due to treatment with immune checkpoint inhibitors are rare but can have life-threatening consequences. If dyspnea and other clinical symptoms are present, AIHA should be considered as a potential cause and treated promptly in a multidisciplinary setting. An expanded comprehension of risk factors and pathogenesis of AIHA is needed to identify high-risk patients beforehand, leading to more effective predictive and reactive treatment approaches.


Subject(s)
Anemia, Hemolytic, Autoimmune , Melanoma , Neoplasms, Second Primary , Humans , Male , Female , Melanoma/drug therapy , Melanoma/etiology , Anemia, Hemolytic, Autoimmune/chemically induced , Anemia, Hemolytic, Autoimmune/therapy , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Immunotherapy/methods , Neoplasms, Second Primary/etiology , Dyspnea/etiology , Adrenal Cortex Hormones/therapeutic use
11.
Leukemia ; 38(6): 1266-1274, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684821

ABSTRACT

Therapy-related myeloid neoplasms (tMN) are complications of cytotoxic therapies. Risk of tMN is high in recipients of autologous hematopoietic stem cell transplantation (aHSCT). Acquisition of genomic mutations represents a key pathogenic driver but the origins, timing and dynamics, particularly in the context of preexisting or emergent clonal hematopoiesis (CH), have not been sufficiently clarified. We studied a cohort of 1507 patients undergoing aHSCT and a cohort of 263 patients who developed tMN without aHSCT to determine clinico-molecular features unique to post-aHSCT tMN. We show that tMN occurs in up to 2.3% of patients at median of 2.6 years post-AHSCT. Age ≥ 60 years, male sex, radiotherapy, high treatment burden ( ≥ 3 lines of chemotherapy), and graft cellularity increased the risk of tMN. Time to evolution and overall survival were shorter in post-aHSCT tMN vs. other tMN, and the earlier group's mutational pattern was enriched in PPM1D and TP53 lesions. Preexisting CH increased the risk of adverse outcomes including post-aHSCT tMN. Particularly, antecedent lesions affecting PPM1D and TP53 predicted tMN evolution post-transplant. Notably, CH-derived tMN had worse outcomes than non CH-derived tMN. As such, screening for CH before aHSCT may inform individual patients' prognostic outcomes and influence their prospective treatment plans. Presented in part as an oral abstract at the 2022 American Society of Hematology Annual Meeting, New Orleans, LA, 2022.


Subject(s)
Clonal Hematopoiesis , Hematopoietic Stem Cell Transplantation , Mutation , Neoplasms, Second Primary , Transplantation, Autologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Middle Aged , Female , Transplantation, Autologous/adverse effects , Adult , Neoplasms, Second Primary/etiology , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/therapy , Aged , Prognosis , Myeloproliferative Disorders/therapy , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Young Adult , Adolescent , Protein Phosphatase 2C/genetics , Tumor Suppressor Protein p53/genetics , Follow-Up Studies , Lymphoma/therapy , Lymphoma/etiology , Lymphoma/genetics , Survival Rate
12.
Zhonghua Xue Ye Xue Za Zhi ; 45(1): 41-47, 2024 Jan 14.
Article in Chinese | MEDLINE | ID: mdl-38527837

ABSTRACT

Objective: To evaluate the efficacy and prognostic factors of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with secondary acute myeloid leukemia (sAML) . Methods: In this multicenter, retrospective clinical study, adult patients aged ≥18 years who underwent allo-HSCT for sAML at four centers of the Zhejiang Hematopoietic Stem Cell Transplantation Collaborative Group from January 2014 to November 2022 were included, and the efficacy and prognostic factors of allo-HSCT were analyzed. Results: A total of 95 patients were enrolled; 66 (69.5%) had myelodysplastic syndrome-acute myeloid leukemia (MDS-AML) , 4 (4.2%) had MDS/MPN-AML, and 25 (26.3%) had therapy-related AML (tAML) . The 3-year CIR, LFS, and overall survival (OS) rates were 18.6% (95% CI 10.2%-27.0%) , 70.6% (95% CI 60.8%-80.4%) , and 73.3% (95% CI 63.9%-82.7%) , respectively. The 3-year CIRs of the M-AML group (including MDS-AML and MDS/MPN-AML) and the tAML group were 20.0% and 16.4%, respectively (P=0.430) . The 3-year LFSs were 68.3% and 75.4%, respectively (P=0.176) . The 3-year OS rates were 69.7% and 75.4%, respectively (P=0.233) . The 3-year CIRs of the groups with and without TP53 mutations were 60.0% and 13.7%, respectively (P=0.003) ; the 3-year LFSs were 20.0% and 76.5%, respectively (P=0.002) ; and the 3-year OS rates were 40.0% and 77.6%, respectively (P=0.002) . According to European LeukmiaNet 2022 (ELN2022) risk stratification, the 3-year CIRs of patients in the low-, intermediate-, and high-risk groups were 8.3%, 17.8%, and 22.6%, respectively (P=0.639) . The three-year LFSs were 91.7%, 69.5%, and 65.6%, respectively (P=0.268) . The 3-year OS rates were 91.7%, 71.4%, and 70.1%, respectively (P=0.314) . Multivariate analysis revealed that advanced disease at allo-HSCT and TP53 mutations were independent risk factors for CIR, LFS, and OS. Conclusion: There was no significant difference in the prognosis of patients who underwent allo-HSCT among the MDS-AML, MDS/MPN-AML, and tAML groups. Advanced disease at transplantation and TP53 mutations were poor prognostic factors. ELN2022 risk stratification had limited value for predicting the prognosis of patients with sAML following allo-HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Neoplasms, Second Primary , Adult , Humans , Adolescent , Prognosis , Retrospective Studies , Myelodysplastic Syndromes/therapy , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , Neoplasms, Second Primary/etiology
13.
Endocrinol Metab (Seoul) ; 39(2): 288-299, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38437824

ABSTRACT

BACKGRUOUND: Current research has not investigated the effect of thyroid-stimulating hormone suppression therapy with levothyroxine on the risk for developing subsequent primary cancers (SPCs). This study aimed to investigate the association between levothyroxine dosage and the risk for SPCs in thyroid cancer patients. METHODS: We conducted a nationwide population-based retrospective cohort study form Korean National Health Insurance database. This cohort included 342,920 thyroid cancer patients between 2004 and 2018. Patients were divided into the non-levothyroxine and the levothyroxine groups, the latter consisting of four dosage subgroups according to quartiles. Cox proportional hazard models were performed to evaluate the risk for SPCs by adjusting for variables including cumulative doses of radioactive iodine (RAI) therapy. RESULTS: A total of 17,410 SPC cases were observed over a median 7.3 years of follow-up. The high-dose levothyroxine subgroups (Q3 and Q4) had a higher risk for SPC (adjusted hazard ratio [HR], 1.14 and 1.27; 95% confidence interval [CI], 1.05-1.24 and 1.17- 1.37; respectively) compared to the non-levothyroxine group. In particular, the adjusted HR of stomach (1.31), colorectal (1.60), liver and biliary tract (1.95), and pancreatic (2.48) cancers were increased in the Q4 subgroup. We consistently observed a positive association between high levothyroxine dosage per body weight and risk of SPCs, even after adjusting for various confounding variables. Moreover, similar results were identified in the stratified analyses according to thyroidectomy type and RAI therapy, as well as in a subgroup analysis of patients with good adherence. CONCLUSION: High-dose levothyroxine use was associated with increased risk of SPCs among thyroid cancer patients regardless of RAI therapy.


Subject(s)
Cancer Survivors , Thyroid Neoplasms , Thyroxine , Humans , Thyroxine/administration & dosage , Thyroxine/therapeutic use , Thyroid Neoplasms/drug therapy , Male , Female , Middle Aged , Retrospective Studies , Adult , Republic of Korea/epidemiology , Cancer Survivors/statistics & numerical data , Aged , Neoplasms, Second Primary/epidemiology , Neoplasms, Second Primary/etiology , Risk Factors , Dose-Response Relationship, Drug , Cohort Studies , Follow-Up Studies
14.
Int J Radiat Oncol Biol Phys ; 119(2): 681-696, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38430101

ABSTRACT

PURPOSE: In this Pediatric Normal Tissue Effects in the Clinic (PENTEC) vision paper, challenges and opportunities in the assessment of subsequent neoplasms (SNs) from radiation therapy (RT) are presented and discussed in the context of technology advancement. METHODS AND MATERIALS: The paper discusses the current knowledge of SN risks associated with historic, contemporary, and future RT technologies. Opportunities for research and SN mitigation strategies in pediatric patients with cancer are reviewed. RESULTS: Present experience with radiation carcinogenesis is from populations exposed during widely different scenarios. Knowledge gaps exist within clinical cohorts and follow-up; dose-response and volume effects; dose-rate and fractionation effects; radiation quality and proton/particle therapy; age considerations; susceptibility of specific tissues; and risks related to genetic predisposition. The biological mechanisms associated with local and patient-level risks are largely unknown. CONCLUSIONS: Future cancer care is expected to involve several available RT technologies, necessitating evidence and strategies to assess the performance of competing treatments. It is essential to maximize the utilization of existing follow-up while planning for prospective data collection, including standardized registration of individual treatment information with linkage across patient databases.


Subject(s)
Cancer Survivors , Neoplasms, Radiation-Induced , Organs at Risk , Humans , Child , Cancer Survivors/statistics & numerical data , Neoplasms, Radiation-Induced/prevention & control , Neoplasms, Radiation-Induced/etiology , Organs at Risk/radiation effects , Proton Therapy/adverse effects , Neoplasms, Second Primary/etiology , Neoplasms, Second Primary/prevention & control , Dose-Response Relationship, Radiation , Dose Fractionation, Radiation , Age Factors , Adolescent , Radiotherapy/adverse effects , Genetic Predisposition to Disease , Neoplasms/radiotherapy
16.
Blood Adv ; 8(11): 2695-2706, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38513082

ABSTRACT

ABSTRACT: Therapy-related myeloid neoplasms (t-MNs) arise after exposure to cytotoxic therapies and are associated with high-risk genetic features and poor outcomes. We analyzed a cohort of patients with therapy-related chronic myelomonocytic leukemia (tCMML; n = 71) and compared its features to that of de novo CMML (dnCMML; n = 461). Median time from cytotoxic therapy to tCMML diagnosis was 6.5 years. Compared with dnCMML, chromosome-7 abnormalities (4% vs 13%; P = .005) but not complex karyotype (3% vs 7%; P = .15), were more frequent in tCMML. tCMML was characterized by higher TP53 mutation frequency (4% vs 12%; P = .04) and lower NRAS (6% vs 22%, P = .007) and CBL (4% vs 12%, P = .04) mutation frequency. Prior therapy with antimetabolites (odd ratio [OR], 1.22; 95% confidence interval [CI], 1.05-1.42; P = .01) and mitotic inhibitors (OR, 1.24; 95% CI, 1.06-1.44; P = .009) was associated with NF1 and SETBP1 mutations whereas prior mitotic inhibitor therapy was associated with lower TET2 mutation frequency (OR, 0.71; 95% CI, 0.55-0.92; P = .01). Although no differences in median overall survival (OS) were observed among tCMML and dnCMML (34.7 months vs 35.9 months, P = .26), multivariate analysis for OS revealed that prior chemotherapy was associated with increased risk of death (hazard ratio, 1.76; 95% CI, 1.07-2.89; P = .026). Compared with a cohort of therapy-related myelodysplastic syndrome, tCMML had lower TP53 mutation frequency (12% vs 44.4%, P < .001) and less unfavorable outcomes. In summary, tCMML does not exhibit the high-risk features and poor outcomes of t-MNs.


Subject(s)
Leukemia, Myelomonocytic, Chronic , Humans , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Male , Female , Aged , Middle Aged , Neoplasms, Second Primary/etiology , Mutation , Aged, 80 and over , Adult , Risk Factors
17.
Blood ; 143(20): 2099-2105, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38483155

ABSTRACT

ABSTRACT: Second primary malignancies were reported in 536 of 12 394 (4.3%) adverse event reports following chimeric antigen receptor T-cell therapies in the Food and Drug Administration Adverse Event Reporting System. Myeloid and T-cell neoplasms were disproportionately more frequently reported, warranting further follow-up.


Subject(s)
Adverse Drug Reaction Reporting Systems , Immunotherapy, Adoptive , Neoplasms, Second Primary , United States Food and Drug Administration , Humans , United States/epidemiology , Neoplasms, Second Primary/etiology , Neoplasms, Second Primary/therapy , Neoplasms, Second Primary/epidemiology , Immunotherapy, Adoptive/adverse effects , Male , Receptors, Chimeric Antigen/immunology , Female , Middle Aged , Adult , Aged
19.
Pediatr Blood Cancer ; 71(6): e30964, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38514796
SELECTION OF CITATIONS
SEARCH DETAIL
...