Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 366
Filter
1.
DNA Repair (Amst) ; 122: 103435, 2023 02.
Article in English | MEDLINE | ID: mdl-36549044

ABSTRACT

New development and optimization of oncologic strategies are steadily increasing the number of long-term cancer survivors being at risk of developing second primary neoplasms (SPNs) as a late consequence of genotoxic cancer therapies with the highest risk among former childhood cancer patients. Since risk factors and predictive biomarkers for therapy-associated SPN remain unknown, we examined the sensitivity to mild replication stress as a driver of genomic instability and carcinogenesis in fibroblasts from 23 long-term survivors of a pediatric first primary neoplasm (FPN), 22 patients with the same FPN and a subsequent SPN, and 22 controls with no neoplasm (NN) using the cytokinesis-block micronucleus (CBMN) assay. Mild replication stress was induced with the DNA-polymerase inhibitor aphidicolin (APH). Fibroblasts from patients with the DNA repair deficiency syndromes Bloom, Seckel, and Fanconi anemia served as positive controls and for validation of the CBMN assay supplemented by analysis of chromosomal aberrations, DNA repair foci (γH2AX/53BP1), and cell cycle regulation. APH treatment resulted in G2/M arrest and underestimation of cytogenetic damage beyond G2, which could be overcome by inhibition of Chk1. Basal micronuclei were significantly increased in DNA repair deficiency syndromes but comparable between NN, FPN, and SPN donors. After APH-induced replication stress, the average yield of micronuclei was significantly elevated in SPN donors compared to FPN (p = 0.013) as well as NN (p = 0.03) donors but substantially lower than for DNA repair deficiency syndromes. Our findings suggest that mild impairment of the response to replication stress induced by genotoxic impacts of DNA-damaging cancer therapies promotes genomic instability in a subset of long-term cancer survivors and may drive the development of an SPN. Our study provides a basis for detailed mechanistic studies as well as predictive bioassays for clinical surveillance, to identify cancer patients at high risk for SPNs at first diagnosis.


Subject(s)
Cancer Survivors , Neoplasms, Second Primary , Humans , Child , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Apoptosis , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Chromosomal Instability , Genomic Instability , Micronucleus Tests/methods , DNA Damage , DNA/metabolism , Fibroblasts/metabolism
2.
Cancer Treat Res Commun ; 31: 100525, 2022.
Article in English | MEDLINE | ID: mdl-35114502

ABSTRACT

INTRODUCTION: Breast cancer (BC) and thyroid dysfunction are common in females, yet the relationship between thyroid hormone and BC is unclear. To search for the connection between thyrotropin and BC, we contradistinguished BC patients with or without synchronous second primary thyroid cancer (TC) with surgery using data from the Surveillance, Epidemiology, and End Results (SEER) database. Theoretically, according to the ATA (American Thyroid Association) guidelines, all TC patients were treated with thyrotropin suppressive therapy only from 2010 to 2015. MATERIALS AND METHODS: Data from BC patients with a synchronous second TC with surgery (BC2TC) and only BC patients (1BC) during 2010-2015 were extracted from the SEER database. Differences in the clinicopathological characteristics between BC2TC and 1BC patients were analyzed by chi-square tests. Comparisons of the disease-specific survival (DSS) and overall survival (OS) curves between these two groups were performed with the log-rank (Mantel-Cox) test. RESULTS: Within this dataset, we identified 134 BC2TC patients during the period from 2010 to 2015. Significant differences between the BC2TC and 1BC groups were found only for different ages and TNM (tumor-node-metastasis status) stages. There were no significant differences in DSS between the two cohorts (P = 0.060). The same tendencies in OS or DSS were observed for the different age groups and different TNM groups, even the stage I, N0 (without metastases to lymph nodes), and ER (+) (estrogen receptor (ER)-positive) groups. CONCLUSIONS: There were no remarkable differences in survival between the BC2TC and 1BC groups, and thyrotropin suppression therapy using levothyroxine did not negatively affect BC prognosis.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Neoplasms, Second Primary , Thyroid Neoplasms , Thyrotropin , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Humans , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/mortality , Neoplasms, Second Primary/pathology , Prognosis , SEER Program , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Thyrotropin/metabolism , Thyroxine/therapeutic use , United States/epidemiology
3.
Blood ; 139(8): 1198-1207, 2022 02 24.
Article in English | MEDLINE | ID: mdl-34469514

ABSTRACT

The BCL2 inhibitor venetoclax has established therapeutic roles in chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML). As BCL2 is an important determinant of survival of both myeloid progenitor and B cells, we investigated whether clinical and molecular abnormalities arise in the myeloid compartment during long-term continuous venetoclax treatment of CLL in 89 patients (87 with relapsed/refractory CLL). Over a median follow-up of 75 (range 21-98) months, persistent cytopenias (≥1 of neutropenia, thrombocytopenia, anemia) lasting ≥4 months and unrelated to CLL occurred in 25 patients (28%). Of these patients, 20 (80%) displayed clonal hematopoiesis, including 10 with therapy-related myeloid neoplasms (t-MNs). t-MNs occurred exclusively in patients previously exposed to fludarabine-alkylator combination therapy with a cumulative 5-year incidence of 10.4% after venetoclax initiation, consistent with rates reported for patients exposed to fludarabine-alkylator combination therapy without venetoclax. To determine whether the altered myelopoiesis reflected the acquisition of mutations, we analyzed samples from patients with no or minimal bone marrow CLL burden (n = 41). Mutations in the apoptosis effector BAX were identified in 32% (13/41). In cellular assays, C-terminal BAX mutants abrogated outer mitochondrial membrane localization of BAX and engendered resistance to venetoclax killing. BAX-mutated clonal hematopoiesis occurred independently of prior fludarabine-alkylator combination therapy exposure and was not associated with t-MNs. Single-cell sequencing revealed clonal co-occurrence of mutations in BAX with DNMT3A or ASXL1. We also observed simultaneous BCL2 mutations within CLL cells and BAX mutations in the myeloid compartment of the same patients, indicating lineage-specific adaptation to venetoclax therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic , Hematologic Neoplasms , Leukemia, Lymphocytic, Chronic, B-Cell , Mutation , Myelopoiesis/drug effects , Myeloproliferative Disorders , Neoplasms, Second Primary , Sulfonamides , bcl-2-Associated X Protein , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Female , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Middle Aged , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Vidarabine/administration & dosage , Vidarabine/adverse effects , Vidarabine/analogs & derivatives , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
6.
Leuk Res ; 111: 106733, 2021 12.
Article in English | MEDLINE | ID: mdl-34749168

ABSTRACT

Recent studies demonstrated that MYC epigenetically regulates AML cell survival and differentiation by suppressing IDH1/2-TET2-5hmC signaling and that MYC overexpression is associated with poor survival outcomes in multiple AML patient cohorts. However, the oncogenic roles of MYC in MDS remain to be explored. A total of 41 patients with de novo MDS were retrospectively identified using the Total Cancer Care database at the Moffitt Cancer Center. A total of 61 % of patients had low MYC expression and 39 % of patients had high MYC expression defined as MYC reactivity by immunohistochemical staining in ≥5% of bone marrow (BM) cells at the time of MDS diagnosis. The median MDS-to-AML progression free survival (PFS) was significantly shorter in the high MYC group (median PFS 9.3 vs. 17.7 months, HR = 2.328, p = 0.013). Further, overall survival (OS) was also shorter in the high MYC patients (median OS 19.7 vs. 51.7 months, HR = 2.299, p = 0.053). Multivariate analyses demonstrated that high MYC expression is an independent poor prognostic factor for the MDS-to-AML progression (HR = 2.275, p = 0.046). Our observations indicate that MYC may play a crucial role in MDS transformation to AML and the underlying mechanisms of MYC-driven MDS clonal expansion and leukemic transformation require further investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/pathology , Mutation , Myelodysplastic Syndromes/pathology , Neoplasms, Second Primary/pathology , Proto-Oncogene Proteins c-myc/metabolism , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Retrospective Studies , Survival Rate , Young Adult
7.
Blood ; 138(9): 790-805, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34473231

ABSTRACT

Therapy-related myeloid neoplasms (t-MNs) are high-risk late effects with poorly understood pathogenesis in cancer survivors. It has been postulated that, in some cases, hematopoietic stem and progenitor cells (HSPCs) harboring mutations are selected for by cytotoxic exposures and transform. Here, we evaluate this model in the context of deficiency of CUX1, a transcription factor encoded on chromosome 7q and deleted in half of t-MN cases. We report that CUX1 has a critical early role in the DNA repair process in HSPCs. Mechanistically, CUX1 recruits the histone methyltransferase EHMT2 to DNA breaks to promote downstream H3K9 and H3K27 methylation, phosphorylated ATM retention, subsequent γH2AX focus formation and propagation, and, ultimately, 53BP1 recruitment. Despite significant unrepaired DNA damage sustained in CUX1-deficient murine HSPCs after cytotoxic exposures, they continue to proliferate and expand, mimicking clonal hematopoiesis in patients postchemotherapy. As a consequence, preexisting CUX1 deficiency predisposes mice to highly penetrant and rapidly fatal therapy-related erythroleukemias. These findings establish the importance of epigenetic regulation of HSPC DNA repair and position CUX1 as a gatekeeper in myeloid transformation.


Subject(s)
Chromosomes, Mammalian , DNA Repair , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Homeodomain Proteins , Leukemia, Erythroblastic, Acute , Neoplasm Proteins , Neoplasms, Second Primary , Nuclear Proteins , Repressor Proteins , Animals , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , Clonal Hematopoiesis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
8.
Int J Mol Sci ; 22(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34208855

ABSTRACT

Patients with advanced colorectal cancer (CRC) with distant metastases have a poor prognosis. We evaluated the clinicopathological relevance of GRP94 expression in these cases. The immunohistochemical expression of GRP94 was studied in 189 CRC patients with synchronous (SM; n = 123) and metachronous metastases (MM; n = 66), using tissue microarray; the association between GRP94 expression, outcome, and tumor-infiltrating lymphocytes (TILs) was also evaluated. GRP94 was expressed in 64.6% (122/189) patients with CRC; GRP94 positivity was found in 67.5% and 59.1% patients with SM and MM, respectively. In the SM group, high GRP94 expression was more common in patients with a higher density of CD4+ TILs (p = 0.002), unlike in the MM group. Survival analysis showed that patients with GRP94 positivity had significantly favorable survival (p = 0.030); after multivariate analysis, GRP94 only served as an independent prognostic factor (p = 0.034; hazard ratio, 0.581; 95% confidence interval, 0.351-0.961) in the SM group. GRP94 expression was detected in 49.4% of metastatic sites and showed significant heterogeneity between primary and metastatic lesions (p = 0.012). GRP94 is widely expressed in CRC with distant metastases; its expression was associated with favorable prognosis in the SM group, unlike in the MM group.


Subject(s)
Colorectal Neoplasms/pathology , Membrane Glycoproteins/metabolism , Neoplasms, Multiple Primary/pathology , Neoplasms, Second Primary/pathology , Up-Regulation , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Neoplasms, Multiple Primary/metabolism , Neoplasms, Second Primary/metabolism , Prognosis , Survival Analysis , Tissue Array Analysis
9.
Br J Cancer ; 125(4): 601-610, 2021 08.
Article in English | MEDLINE | ID: mdl-34040177

ABSTRACT

BACKGROUND: We sought to estimate the annual risk and 25-year cumulative risk of contralateral breast cancer among women with stage 0-III unilateral breast cancer. METHODS: We identified 812,851 women with unilateral breast cancer diagnosed between 1990 and 2015 in the SEER database and followed them for contralateral breast cancer for up to 25 years. Women with a known bilateral mastectomy were excluded. We calculated the annual risk of contralateral breast cancer by age at diagnosis, by time since diagnosis and by current age. We compared risks by ductal carcinoma in situ (DCIS) versus invasive disease, by race and by oestrogen receptor (ER) status of the first cancer. RESULTS: There were 25,958 cases of contralateral invasive breast cancer diagnosed (3.2% of all patients). The annual risk of contralateral breast cancer over the 25-year follow-up period was 0.37% and the 25-year actuarial risk of contralateral invasive breast cancer was 9.9%. The annual risk varied to a small degree by age of diagnosis, by time elapsed since diagnosis and by current age. The 25-year actuarial risk was similar for DCIS and invasive breast cancer patients (10.1 versus 9.9%). The 25-year actuarial risk was higher for black women (12.7%) than for white women (9.7%) and was lower for women with ER-positive breast cancer (9.5%) than for women with ER-negative breast cancer (11.2%). CONCLUSIONS: Women with unilateral breast cancer experience an annual risk of contralateral breast cancer ~0.4% per year, which persists over the 25-year follow-up period.


Subject(s)
Breast Neoplasms/epidemiology , Carcinoma, Ductal, Breast/epidemiology , Neoplasms, Second Primary/epidemiology , Adult , Age Factors , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Female , Humans , Middle Aged , Neoplasm Staging , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology , Receptors, Estrogen/metabolism , Risk Factors , SEER Program
10.
Blood ; 138(9): 749-757, 2021 09 02.
Article in English | MEDLINE | ID: mdl-33876223

ABSTRACT

Therapy-related myeloid neoplasms (t-MNs) include diseases onsetting in patients treated with chemo- and/or radiotherapy for a primary cancer, or an autoimmune disorder. Genomic variants, in particular, in familial cancer genes, may play a predisposing role. Recent advances in deep sequencing techniques have shed light on the pathogenesis of t-MNs, identifying clonal hematopoiesis of indeterminate potential (CHIP) as a frequent first step in the multihit model of t-MNs. CHIP is often detectable prior to any cytotoxic treatment, probably setting the fertile genomic background for secondary leukemogenesis. The evolution pattern toward t-MNs is then a complex process, shaped by the type of cancer therapy, the aging process, and the individual exposures, that favor additional hits, such as the acquisition of TP53 mutations and unfavorable karyotype abnormalities. The pathogenesis of t-MNs differs from MN associated with environmental exposure. Indeed, the genetic aberration patterns of MN developing in atomic bomb survivors show few mutations in classical DNA methylation genes, and a high prevalence of 11q and ATM alterations, together with TP53 mutations. Survival in t-MNs is poor. In addition to the biology of t-MNs, the patient's previous disease history and the remission status at t-MN diagnosis are significant factors contributing to unfavorable outcome. New drugs active in secondary leukemias include CPX-351, or venetoclax in combination with hypomethylating agents, monoclonal antibodies as magrolimab, or targeted drugs against pathogenic mutations. Allogeneic stem cell transplantation remains the best currently available therapeutic option with curative intent for fit patients with unfavorable genetic profiles.


Subject(s)
Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Mutation , Myeloproliferative Disorders , Neoplasm Proteins , Neoplasms, Second Primary , Allografts , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology , Neoplasms, Second Primary/therapy
11.
Breast ; 57: 71-79, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33774461

ABSTRACT

BACKGROUND: The aim of this study was to analyze the association of molecular subtype concordance and disease outcome in patients with synchronous bilateral breast cancer (SBBC) and metachronous breast cancer (MBBC). PATIENTS AND METHODS: Patients diagnosed with SBBC or MBBC in the Surveillance, Epidemiology, and End Results (SEER) database or Comprehensive Breast Health Center (CBHC) Ruijin Hospital, Shanghai were retrospectively reviewed and included. Clinicopathologic features, molecular subtype status concordance, and prognosis were compared in patients with SBBC and MBBC. Other prognostic factors for breast cancer-specific survival (BCSS) and overall survival (OS) were also identified for bilateral breast cancer patients. RESULTS: Totally, 3395 and 115 patients were included from the SEER and Ruijin CBHC cohorts. Molecular subtype concordance rate was higher in the SBBC group compared to MBBC in both SEER cohort (75.8% vs 57.7%, p < 0.001) and Ruijin CBHC cohort (76.2% vs 45.2%, p = 0.002). Survival analyses indicated that SBBC was related to worse BCSS than MBBC (p = 0.015). Molecular subtype discordance was related to worse BCSS (hazard ratio (HR), 1.64, 95% confidential interval (CI), 1.18-2.27, p = 0.003) and OS (HR, 1.59, 95% CI, 1.24-2.04, p < 0.001) in the SBBC group, but not for the MBBC group (p = 0.650 for BCSS, p = 0.669 for OS). CONCLUSIONS: Molecular subtype concordance rate was higher in the SBBC group than MBBC group. Patients with discordant molecular subtype was associated with worse disease outcome in the SBBC patients, but not in MBBC, which deserves further clinical evaluation.


Subject(s)
Breast Neoplasms/pathology , Neoplasms, Multiple Primary/pathology , Neoplasms, Second Primary/mortality , Neoplasms, Second Primary/pathology , Adult , Aged , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , China/epidemiology , Female , Humans , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasms, Multiple Primary/mortality , Neoplasms, Second Primary/metabolism , Population Surveillance , Prognosis , Retrospective Studies
14.
J Cutan Pathol ; 48(4): 479-485, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32939793

ABSTRACT

BACKGROUND: PRAME (preferentially expressed antigen in melanoma) is a promising immunohistochemical marker in distinguishing benign from malignant primary cutaneous melanocytic lesions and lymph node deposits. We hypothesize that PRAME may also reliably identify melanoma metastases that are clinically detected in skin, lymph nodes, or small intestine. METHODS: A total of 155 cases of metastatic melanoma to lymph node (N = 54) and non-lymph node (N = 101) sites were stained with an antibody against PRAME. Nuclear expression was scored in tumor cells as negative, 1% to 25% (1+), 26% to 50% (2+), 51% to 75% (3+), or 76% to 100% (4+). RESULTS: PRAME expression was seen in 151/155 (97.4%) cases, with 4+ expression in 64 cases (41.3%), 3+ expression in 46 cases (29.7%), 2+ expression in 18 cases (11.6%), and 1+ expression in 23 cases (14.8%). Lymph node metastases were more likely to show lower expression as compared to metastases to other anatomic sites (P = 0.003). CONCLUSIONS: A high level of PRAME immunoreactivity was identified in this cohort of metastatic melanoma. Lymph node metastases showed more focal or absent PRAME expression as compared to metastases to other sites. Overall, PRAME is a useful tool for confirming the diagnosis of melanoma in a metastatic setting, in both nodal and visceral deposits.


Subject(s)
Antigens, Neoplasm/metabolism , Immunohistochemistry/methods , Melanoma/diagnosis , Neoplasms, Second Primary/metabolism , Skin Neoplasms/pathology , Aged, 80 and over , Female , Humans , Immunotherapy/methods , Intestine, Small/pathology , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Male , Melanoma/secondary , Middle Aged , Neoplasms, Second Primary/pathology
15.
Urol Int ; 105(1-2): 21-26, 2021.
Article in English | MEDLINE | ID: mdl-33049748

ABSTRACT

BACKGROUND: MicroRNA-371a-3p (miR-371), the novel serum biomarker of testicular germ cell tumours (GCTs), is produced by undifferentiated subtypes of GCTs but not by teratoma. Cystic teratoma developing from retroperitoneal metastases of GCT subsequent to chemotherapy had been shown to contain high levels of classical serum tumour markers of GCT in the presence of normal marker levels in serum. To date, no information is available regarding the presence of miR-371 in the cystic fluid of residual teratoma after chemotherapy. METHODS: Four patients (age 18-26 years) undergoing retroperitoneal lymph node dissection (RPLND) for cystic residual masses resulting from chemotherapy of bulky retroperitoneal GCT had measurements of miR-371 in both serum and cystic fluid aspirated from surgical specimens. Measurement of the miR was performed with quantitative real-time PCR using miR-30b-5p as reference. Results were tabulated and analysed in a descriptive manner. RESULTS: Histologically, all of the surgical specimens involved teratoma only with no evidence of vital undifferentiated GCT tissue. All patients were cured. Prior to RPLND, miR-371 serum levels were not measurable or close to zero in all of the patients. Cystic fluid revealed elevated levels of miR-371 in 3 patients and traces of miR in one. CONCLUSIONS: The detection of miR-371 in the cystic fluid of teratoma is somewhat enigmatic since this GCT subtype usually does not express the miR. Two hypotheses may explain the finding: First, miR-371 molecules were released into the cystic fluid by active GCT tissue prior to chemotherapy. High levels were kept after regression of vital GCT tissue because the cystic lumen is without a specific drainage system. Second, teratoma cells lining the interior cyst wall may shed small amounts of miR-371 into the lumen. Because of the lacking drainage system, even small levels may accumulate. The present finding adds to the understanding of the biology of the novel biomarker of GCT.


Subject(s)
Biomarkers, Tumor/analysis , Cyst Fluid/chemistry , MicroRNAs/analysis , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Second Primary/metabolism , Retroperitoneal Neoplasms/metabolism , Teratoma/metabolism , Testicular Neoplasms/drug therapy , Adolescent , Adult , Biomarkers, Tumor/biosynthesis , Humans , Male , MicroRNAs/biosynthesis , Young Adult
16.
Oral Oncol ; 114: 105089, 2021 03.
Article in English | MEDLINE | ID: mdl-33189577

ABSTRACT

It is well known that head and neck squamous cell carcinomas are characterized by genetic alterations, genomic instability and different immune defects and Telomerase reverse transcriptase promoter mutations have been found with high rates in patients with tongue squamous cell carcinomas. We presented a non-smoker, no alcohol consuming woman who had a second primary tongue SCC after four and a half years later of her first tongue SCC with TERT promoter mutation. The primary tumor and its recurrences in woman with tongue SCC who have never smoked and had no alcohol consumption may need to be differing from other tongue carcinomas. TERT promoter mutations may have a relation in etiopathogenesis of tumor and mutations need to be on mind in future studies.


Subject(s)
Mutation , Neoplasms, Second Primary/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Telomerase/genetics , Tongue Neoplasms/genetics , Aged, 80 and over , Female , Humans , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology , Promoter Regions, Genetic , Squamous Cell Carcinoma of Head and Neck/metabolism , Telomerase/metabolism , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology
18.
Leuk Res ; 98: 106453, 2020 11.
Article in English | MEDLINE | ID: mdl-33059120

ABSTRACT

We described the clinical features and outcomes for 63 adult patients with acute myeloid leukemia (AML) with a translocation involving the 11q23 locus (MLL) who were treated at Memorial Sloan Kettering Cancer Center (MSK). The population included 40 female (63 %) and 23 male (37 %) patients, with a median age of 51 years old (range 18-82 years). Of the 31 patients who had had an antecedent malignancy, 14 (45 %) had had breast cancer or DCIS and 22 (71 %) had received anthracycline-based systemic chemotherapy. The translocation partner for the 11q23 rearrangement was identified in 60 of the 63 patients (95 %) studied. The distribution of translocation partners differed for those who had previously received cytotoxic chemotherapy. Most patients with therapy-related disease had a 9p22 or 19p13 partner, as compared to those with de novo disease (95 % vs. 68 %, p = 0.023). Of the 30 patients who received all therapy under observation, 15 (50 %) patients had de novo disease and 15 (50 %) had received antecedent chemotherapy. No significant difference in survival was observed between groups (p = 0.44). Twenty-two patients received induction as up-front therapy, of whom 11 (50 %) achieved CR / CRi. The achievement of CR / CRi with one course of induction was associated with improved OS, with a 6-month OS of 73 % as compared to 23 % for those who did not (p = 0.018). The achievement of CR / CRi with a single course of induction may be a marker of favorable survival in this subtype of high-risk AML. KEY POINT: Response to a single induction was associated with favorable survival in this population.


Subject(s)
Anthracyclines/administration & dosage , Breast Neoplasms , Chromosomes, Human, Pair 11 , Leukemia, Myeloid, Acute , Neoplasms, Second Primary , Translocation, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/metabolism , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 19/metabolism , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology
19.
Clin Nucl Med ; 45(12): 1013-1015, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33086276

ABSTRACT

A bone lesion in the right calcaneus was found in a 76-year-old man with a history of prostate cancer. Ga-PSMA-11 PET/CT showed multiple PSMA-expressing lytic bone lesions in appendicular skeleton. Biopsy of the lesion in the right calcaneus confirmed plasmacytoma. After several cycles of chemotherapy, the patient was in serologically stringent complete remission. The follow-up PET/CT showed a significant reduction of Ga-PSMA-11 uptake in the previously PSMA-avid bone lesions without morphological changes on CT. The reduction of PSMA expression in myelomas after chemotherapy is suggested to result from the treatment response.


Subject(s)
Membrane Glycoproteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neoplasms, Second Primary/metabolism , Organometallic Compounds/metabolism , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/metabolism , Aged , Biological Transport , Gallium Isotopes , Gallium Radioisotopes , Humans , Male , Multiple Myeloma/diagnostic imaging
20.
Clin Exp Metastasis ; 37(6): 649-656, 2020 12.
Article in English | MEDLINE | ID: mdl-33099724

ABSTRACT

Colorectal cancer (CRC) patients suffer from the second highest mortality among all cancer entities. In half of all CRC patients, colorectal cancer liver metastases (CRLM) can be observed. Metastatic colorectal cancer is associated with poor overall survival and limited treatment options. Even after successful surgical resection of the primary tumor, metachronous liver metastases occur in one out of eight cases. The only available curative intended treatment is hepatic resection, but metachronous CRLM frequently recur after approximately 1 year. In this study, we performed a proteome analysis of three recurrent liver metastases of a single CRC patient by mass spectrometry. Despite surgical resection of the primary CRC and adjuvant chemotherapy plus cetuximab treatment, the patient developed three metachronous CRLM which occurred consecutively after 9, 21 and 31 months. We identified a set of 1132 proteins expressed in the three metachronous CRLM, of which 481 were differentially regulated, including 81 proteins that were associated with the extracellular matrix (ECM). 56 ECM associated proteins were identified as upregulated in the third metastasis, 26 (46%) of which were previously described as negative prognostic markers in CRC, including tenascin C, nidogen 1, fibulin 1 and vitronectin. These data may reflect an ascending trend of malignancy from the first to the third metachronous colorectal cancer liver metastasis. Additionally, the results indicate different ECM phenotypes for recurrent metachronous metastasis, associated with different grades of malignancy and highlights the importance of individual analysis of molecular features in different, consecutive metastatic events in a single patient.


Subject(s)
Colorectal Neoplasms/metabolism , Extracellular Matrix Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Liver Neoplasms/drug therapy , Male , Mass Spectrometry , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology , Organoplatinum Compounds/administration & dosage , Proteome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...