Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Cancer Immunol Immunother ; 71(3): 675-688, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34342668

ABSTRACT

Many nasopharyngeal carcinoma (NPC) patients develop distant metastases after treatment, leading to poor outcomes. To date, there are no peripheral biomarkers suitable for all NPC patients to predict distant metastasis. Hence, we purposed to develop a noninvasive comprehensive model for predicting post-treatment distant metastasis of all NPC. Since T-cell receptor ß chain (TCRB) repertoire has achieved prognostic prediction in many cancers, the clinical characteristics and parameters of TCRB repertoire of 71 cases of peripheral blood samples (pairwise pre-treatment and post-treatment samples from 40 NPC patients who without (nM, n = 21) or with (M, n = 19) post-treatment distant metastasis) were collected. The least absolute shrinkage and selection operator algorithm was used to construct a distant metastasis prediction model. In terms of TCRB repertoire parameters, the diversity of TCRB repertoire was significantly decreased in M group after treatment but not in nM group. Ascending TCRB diversity and higher similarity between pre- and post-treatment samples showed better distant metastasis-free survival (DMFS). The similarity still had robust DMFS prediction in patients with reduced TCRB diversity. More importantly, the 5-factor comprehensive model consisting of basic clinical characteristics and TCRB repertoire indices showed a higher prognostic accuracy than any one individual factor in DMFS predicting. In conclusion, treatment had different effects on the composition of TCRB repertoire in patients without and with post-treatment distant metastasis. The dynamics of TCRB diversity, the similarity of TCRB repertoires, and combinations of these factors with basic clinical characteristics could serve as noninvasive DMFS predictors for all NPC patients.


Subject(s)
Biomarkers , Models, Biological , Nasopharyngeal Carcinoma/diagnosis , Nasopharyngeal Carcinoma/etiology , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/etiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Clonal Evolution/drug effects , Clonal Evolution/genetics , Computational Biology/methods , Disease Progression , Gene Expression Profiling , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Leukocyte Count , Lymphocyte Count , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/therapy , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/pathology
2.
Cancer Res ; 81(19): 4926-4938, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34413060

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) often recurs after chemoradiotherapy, and the prognosis of ESCC after chemoradiotherapy has not improved over the past few decades. The mutation process in chemoradiotherapy-resistant clones and the functional relevance of genetic alterations remain unclear. To address these problems, we performed whole-exome sequencing of 52 tumor samples from 33 patients with ESCC who received radiotherapy combined with 5-fluorouracil/platinum. In multiregion analyses of pretreatment and locally recurrent lesions from five cases, most driver gene-altered clones remained under chemoradiotherapy selection pressure, while few driver gene alterations were acquired at recurrence. The mutation signatures of recurrent ESCC, including increased deletion frequency and platinum dose-dependent base substitution signatures, were substantially different from those of primary ESCC and reflected the iatrogenic impacts of chemoradiotherapy. Single-region analysis of 28 pretreatment tumors indicated that focal copy-number gain at the MYC locus was significantly associated with poor progression-free survival and overall survival after chemoradiotherapy. MYC gain remained throughout the chemoradiotherapy course and potentially contributes to intrinsic resistance to chemoradiotherapy. Consistent with these findings, MYC copy number and mRNA and protein levels in ESCC cell lines correlated positively with resistance to radiotherapy, and MYC knockdown improved sensitivity to radiotherapy. Overall, these data characterize the clonal evolution process induced by chemoradiotherapy and clinically relevant associations for genetic alterations in ESCC. These findings increase our understanding of therapeutic resistance and support the rationale for precision chemoradiotherapy. SIGNIFICANCE: Whole-exome sequencing reveals the genetic evolution of ESCC during chemoradiotherapy, highlighting MYC gain in pretreatment tumors as a potential marker of therapy resistance.


Subject(s)
Biomarkers, Tumor , Esophageal Squamous Cell Carcinoma/genetics , Evolution, Molecular , Genomics , Chemoradiotherapy , Clonal Evolution/drug effects , Clonal Evolution/genetics , Clonal Evolution/radiation effects , Computational Biology/methods , Databases, Genetic , Disease Management , Drug Resistance, Neoplasm/genetics , Esophageal Squamous Cell Carcinoma/diagnosis , Esophageal Squamous Cell Carcinoma/therapy , Genetic Predisposition to Disease , Genomics/methods , Humans , INDEL Mutation , Mutation , Neoplasm Recurrence, Local , Neoplasm Staging , Polymorphism, Single Nucleotide , Prognosis , Radiation Tolerance/genetics , Tumor Burden , Exome Sequencing
3.
Nat Commun ; 12(1): 4803, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376657

ABSTRACT

Chemotherapies may increase mutagenesis of healthy cells and change the selective pressures in tissues, thus influencing their evolution. However, their contributions to the mutation burden and clonal expansions of healthy somatic tissues are not clear. Here, exploiting the mutational footprint of some chemotherapies, we explore their influence on the evolution of hematopoietic cells. Cells of Acute Myeloid Leukemia (AML) secondary to treatment with platinum-based drugs show the mutational footprint of these drugs, indicating that non-malignant blood cells receive chemotherapy mutations. No trace of the 5-fluorouracil (5FU) mutational signature is found in AMLs secondary to exposure to 5FU, suggesting that cells establishing the leukemia could be quiescent during treatment. Using the platinum-based mutational signature as a barcode, we determine that the clonal expansion originating the secondary AMLs begins after the start of the cytotoxic treatment. Its absence in clonal hematopoiesis cases is consistent with the start of the clonal expansion predating the exposure to platinum-based drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Hematopoiesis/drug effects , Leukemia, Myeloid/genetics , Mutagenesis/drug effects , Acute Disease , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Clonal Evolution/drug effects , Clonal Evolution/genetics , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , Cohort Studies , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Hematopoiesis/genetics , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid/chemically induced , Mutation/drug effects , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/genetics , Platinum/administration & dosage , Platinum/adverse effects , Tumor Suppressor Protein p53/genetics
4.
Blood ; 138(23): 2372-2382, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34314481

ABSTRACT

Complex karyotype, defined as ≥3 cytogenetic abnormalities, is prognostic of survival in patients treated with ibrutinib or venetoclax in relapsed/refractory (RR) chronic lymphocytic leukemia (CLL). Recent studies re-evaluating this dichotomous variable have shown that higher numbers of cytogenetic abnormalities (ie, ≥5) have a worse overall survival in patients treated with chemoimmunotherapy. We sought to determine if increasing karyotypic complexity, treated as a continuous variable, was prognostic of survival for patients treated with ibrutinib for CLL. We conducted a retrospective analysis of all patients with CLL treated with single-agent ibrutinib or in combination with an anti-CD20 antibody at our institution. We included 456 patients with both treatment-naive and RR disease. Median number of prior therapies was 2 (range, 0-13), 30% of patients had presence of del(17p), and 75% expressed unmutated IGHV. Fifty percent had ≥3 cytogenetic abnormalities, including 30% with ≥5. In a multivariable analysis, increasing karyotypic complexity was an independent predictor of shorter progression-free survival (hazard ratio, 1.07; 95% confidence interval, 1.04-1.10; P < .0001) and overall survival (hazard ratio, 1.09; 95% confidence interval, 1.05-1.12; P < .0001). Furthermore, we found that presence of clonal evolution determined by cytogenetic analysis at progression was prognostic of subsequent survival (P = .02). This solidifies karyotypic complexity as an important prognostic factor for patients with CLL treated with ibrutinib. Further research should consider sequential karyotypic analysis as a determination of risk of progression and death in patients with CLL.


Subject(s)
Abnormal Karyotype , Adenine/analogs & derivatives , Clonal Evolution , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Abnormal Karyotype/drug effects , Adenine/therapeutic use , Adult , Aged , Aged, 80 and over , Clonal Evolution/drug effects , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Male , Middle Aged , Prognosis , Progression-Free Survival , Retrospective Studies , Survival Analysis
5.
Adv Cancer Res ; 151: 39-67, 2021.
Article in English | MEDLINE | ID: mdl-34148619

ABSTRACT

Multi-modal treatment of non-metastatic locally advanced rectal adenocarcinoma (LARC) includes chemotherapy, radiation, and life-altering surgery. Although highly effective for local cancer control, metastatic failure remains significant and drives rectal cancer-related mortality. A consistent observation of this tri-modality treatment paradigm is that histologic response of the primary tumor to neoadjuvant treatment(s), which varies across patients, predicts overall oncologic outcome. In this chapter, we will examine this treatment response heterogeneity in the context of evolutionary dynamics. We hypothesize that improved understanding of eco-evolutionary pressures rendering small cancer cell populations vulnerable to extinction may influence treatment strategies and improve patient outcomes. Applying effective treatment(s) to cancer populations causes a "race to extinction." We explore principles of eco-evolutionary extinction in the context of these small cancer cell populations, evaluating how treatment(s) aim to eradicate the cancer populations to ultimately result in cure. In this chapter, we provide an evolutionary rationale for limiting continuous treatment(s) with the same agent or combination of agents to avoid selection of resistant cancer subpopulation phenotypes, allowing "evolutionary rescue." We draw upon evidence from nature demonstrating species extinction rarely occurring as a single event phenomenon, but rather a series of events in the slide to extinction. We posit that eradicating small cancer populations, similar to small populations in natural extinctions, will usually require a sequence of different external perturbations that produce negative, synergistic dynamics termed the "extinction vortex." By exploiting these unique extinction vulnerabilities of small cancer populations, the optimal therapeutic sequences may be informed by evolution-informed strategies for patients with LARC.


Subject(s)
Adenocarcinoma/pathology , Clonal Evolution/physiology , Neoadjuvant Therapy/adverse effects , Rectal Neoplasms/pathology , Adaptation, Physiological/drug effects , Adaptation, Physiological/radiation effects , Adenocarcinoma/therapy , Animals , Chemotherapy, Adjuvant/adverse effects , Clonal Evolution/drug effects , Clonal Evolution/radiation effects , Disease Progression , Humans , Radiotherapy/adverse effects , Rectal Neoplasms/therapy
6.
Blood ; 138(25): 2670-2685, 2021 12 23.
Article in English | MEDLINE | ID: mdl-33945616

ABSTRACT

Patients with chronic lymphocytic leukemia (CLL) bearing TP53 mutations experience chemorefractory disease and are therefore candidates for targeted therapy. However, the significance of low-burden TP53 mutations with <10% variant allele frequency (VAF) remains a matter for debate. Herein, we describe clonal evolution scenarios of low-burden TP53 mutations, the clinical impact of which we analyzed in a "real-world" CLL cohort. TP53 status was assessed by targeted next-generation sequencing (NGS) in 511 patients entering first-line treatment with chemo- and/or immunotherapy and 159 patients in relapse before treatment with targeted agents. Within the pretherapy cohort, 16% of patients carried low-burden TP53 mutations (0.1% to 10% VAF). Although their presence did not significantly shorten event-free survival after first-line therapy, it affected overall survival (OS). In a subgroup with TP53 mutations of 1% to 10% VAF, the impact on OS was observed only in patients with unmutated IGHV who had not received targeted therapy, as patients benefited from switching to targeted agents, regardless of initial TP53 mutational status. Analysis of the clonal evolution of low-burden TP53 mutations showed that the highest expansion rates were associated with fludarabine, cyclophosphamide, and rituximab regimen in both first- and second-line treatments (median VAF increase, 14.8× and 11.8×, respectively) in contrast to treatment with less intense treatment regimens (1.6×) and no treatment (0.8×). In the relapse cohort, 33% of patients carried low-burden TP53 mutations, which did not expand significantly upon targeted treatment (median VAF change, 1×). Sporadic cases of TP53 mutations' clonal shifts were connected with the development of resistance-associated mutations. Altogether, our data support the incorporation of low-burden TP53 variants in clinical decision making.


Subject(s)
Clonal Evolution , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clonal Evolution/drug effects , Female , Humans , Immunotherapy , Kaplan-Meier Estimate , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Male , Middle Aged , Mutation/drug effects , Tumor Cells, Cultured
7.
Blood ; 137(22): 3093-3104, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33598693

ABSTRACT

In the international randomized phase 3 RATIFY (Randomized AML Trial In FLT3 in patients less than 60 Years old) trial, the multikinase inhibitor midostaurin significantly improved overall and event-free survival in patients 18 to 59 years of age with FLT3-mutated acute myeloid leukemia (AML). However, only 59% of patients in the midostaurin arm achieved protocol-specified complete remission (CR), and almost half of patients achieving CR relapsed. To explore underlying mechanisms of resistance, we studied patterns of clonal evolution in patients with FLT3-internal tandem duplications (ITD)-positive AML who were entered in the RATIFY or German-Austrian Acute Myeloid Leukemia Study Group 16-10 trial and received treatment with midostaurin. To this end, paired samples from 54 patients obtained at time of diagnosis and at time of either relapsed or refractory disease were analyzed using conventional Genescan-based testing for FLT3-ITD and whole exome sequencing. At the time of disease resistance or progression, almost half of the patients (46%) became FLT3-ITD negative but acquired mutations in signaling pathways (eg, MAPK), thereby providing a new proliferative advantage. In cases with FLT3-ITD persistence, the selection of resistant ITD clones was found in 11% as potential drivers of disease. In 32% of cases, no FLT3-ITD mutational change was observed, suggesting either resistance mechanisms bypassing FLT3 inhibition or loss of midostaurin inhibitory activity because of inadequate drug levels. In summary, our study provides novel insights into the clonal evolution and resistance mechanisms of FLT3-ITD-mutated AML under treatment with midostaurin in combination with intensive chemotherapy.


Subject(s)
Clonal Evolution/drug effects , Leukemia, Myeloid, Acute , Mutation , Staurosporine/analogs & derivatives , fms-Like Tyrosine Kinase 3 , Adolescent , Adult , Aged , Clonal Evolution/genetics , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Staurosporine/administration & dosage , Tandem Repeat Sequences , Exome Sequencing , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
9.
Biochem Biophys Res Commun ; 534: 461-467, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33246559

ABSTRACT

Treatment relapse due to clonal evolution was shown to be an independent factor for poor prognosis in advanced stages of chronic myeloid leukemia. Overcoming secondary resistance arising due to clonal evolution is still an unmet need and lack of adequate pre-clinical models hampers the identification of underlying mechanisms and testing of alternate treatment strategies. The current study thus aimed to create cellular models to study molecular mechanisms underlying clonal evolution and identify strategies to overcome the secondary drug resistance. Analysis of cell lines derived from three independent cell-based screens revealed the co-evolution specifically of imatinib and HSP90 inhibitor (HSP90i) resistances despite their exposure to a single inhibitor alone. Molecular and biochemical characterization of these cell lines revealed additional cytogenetic abnormalities, differential activation of pro-survival signaling molecules and over expression of ABL kinase and HSP90 genes. Importantly, all the imatinib-HSP90i dual resistant cell lines remained sensitive to sorafenib and vorinostat suggesting their utility in treating patients who relapse upon imatinib treatment due to clonal evolution. In addition, we cite similar examples of dual resistance towards various kinase inhibitors and HSP90i in some cell lines that represent solid cancers suggesting co-evolution leading to secondary drug resistance as a pan-cancer phenomenon. Taken together, our results suggest the efficacy of HSP90i in overcoming drug resistance caused by point mutations in the target kinase but not in cases of clonal evolution.


Subject(s)
Antineoplastic Agents/pharmacology , Clonal Evolution/drug effects , Drug Resistance, Neoplasm , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Cell Line, Tumor , Chromosome Aberrations/drug effects , HSP90 Heat-Shock Proteins/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Transcriptome/drug effects
10.
Nat Commun ; 11(1): 5327, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087716

ABSTRACT

Clonal diversity is a consequence of cancer cell evolution driven by Darwinian selection. Precise characterization of clonal architecture is essential to understand the evolutionary history of tumor development and its association with treatment resistance. Here, using a single-cell DNA sequencing, we report the clonal architecture and mutational histories of 123 acute myeloid leukemia (AML) patients. The single-cell data reveals cell-level mutation co-occurrence and enables reconstruction of mutational histories characterized by linear and branching patterns of clonal evolution, with the latter including convergent evolution. Through xenotransplantion, we show leukemia initiating capabilities of individual subclones evolving in parallel. Also, by simultaneous single-cell DNA and cell surface protein analysis, we illustrate both genetic and phenotypic evolution in AML. Lastly, single-cell analysis of longitudinal samples reveals underlying evolutionary process of therapeutic resistance. Together, these data unravel clonal diversity and evolution patterns of AML, and highlight their clinical relevance in the era of precision medicine.


Subject(s)
Clonal Evolution/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Aged , Animals , Clonal Evolution/drug effects , Cohort Studies , Female , Genetic Association Studies , Genomics , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Mice , Mice, Inbred NOD , Middle Aged , Models, Genetic , Mutation , Sequence Analysis, DNA , Single-Cell Analysis
11.
Blood ; 136(14): 1599-1605, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32736382

ABSTRACT

Clonal hematopoiesis (CH) is common in older persons and is associated with an increased risk of hematologic cancer. Here, we review studies establishing an association between CH and hematopoietic malignancy, discuss features of CH that are predictive of leukemic progression, and explore the role of hematopoietic stressors in the evolution of CH to acute myeloid leukemia or myelodysplastic syndrome. CH due to point mutations or structural variants such as copy-number alterations is associated with an ∼10-fold increased risk of hematopoietic malignancy. Although the absolute risk of hematopoietic malignancy is low, certain features of CH may confer a higher risk of transformation, including the presence of TP53 or spliceosome gene mutations, a variant allele fraction >10%, the presence of multiple mutations, and altered red blood indices. CH in the setting of peripheral blood cytopenias carries a very high risk of progression to a myeloid malignancy and merits close observation. There is emerging evidence suggesting that hematopoietic stressors contribute to both the development of CH and progression to hematopoietic malignancy. Specifically, there is evidence that genotoxic stress from chemotherapy or radiation therapy, ribosome biogenesis stress, and possibly inflammation may increase the risk of transformation from CH to a myeloid malignancy. Models that incorporate features of CH along with an assessment of hematopoietic stressors may eventually help predict and prevent the development of hematopoietic malignancies.


Subject(s)
Clonal Hematopoiesis , Disease Susceptibility , Hematologic Neoplasms/etiology , Hematopoiesis , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Biomarkers , Cell Transformation, Neoplastic/genetics , Clonal Evolution/drug effects , Clonal Evolution/genetics , Clonal Hematopoiesis/drug effects , Clonal Hematopoiesis/genetics , Genetic Predisposition to Disease , Hematopoiesis/drug effects , Hematopoiesis/genetics , Humans , Mutation , Neoplasms, Second Primary/etiology , Pancytopenia/etiology , Stress, Physiological
12.
Clin Cancer Res ; 26(18): 4958-4969, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32616500

ABSTRACT

PURPOSE: Using next-generation sequencing (NGS), we recently documented T-cell oligoclonality in treatment-naïve chronic lymphocytic leukemia (CLL), with evidence indicating T-cell selection by restricted antigens. EXPERIMENTAL DESIGN: Here, we sought to comprehensively assess T-cell repertoire changes during treatment in relation to (i) treatment type [fludarabine-cyclophosphamide-rituximab (FCR) versus ibrutinib (IB) versus rituximab-idelalisib (R-ID)], and (ii) clinical response, by combining NGS immunoprofiling, flow cytometry, and functional bioassays. RESULTS: T-cell clonality significantly increased at (i) 3 months in the FCR and R-ID treatment groups, and (ii) over deepening clinical response in the R-ID group, with a similar trend detected in the IB group. Notably, in constrast to FCR that induced T-cell repertoire reconstitution, B-cell receptor signaling inhibitors (BcRi) preserved pretreatment clones. Extensive comparisons both within CLL as well as against T-cell receptor sequence databases showed little similarity with other entities, but instead revealed major clonotypes shared exclusively by patients with CLL, alluding to selection by conserved CLL-associated antigens. We then evaluated the functional effect of treatments on T cells and found that (i) R-ID upregulated the expression of activation markers in effector memory T cells, and (ii) both BcRi improved antitumor T-cell immune synapse formation, in marked contrast to FCR. CONCLUSIONS: Taken together, our NGS immunoprofiling data suggest that BcRi retain T-cell clones that may have developed against CLL-associated antigens. Phenotypic and immune synapse bioassays support a concurrent restoration of functionality, mostly evident for R-ID, arguably contributing to clinical response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Clonal Evolution/drug effects , Immunological Synapses/drug effects , Leukemia, Prolymphocytic, T-Cell/drug therapy , T-Lymphocytes/drug effects , Adenine/administration & dosage , Adenine/analogs & derivatives , Aged , Aged, 80 and over , Clonal Evolution/immunology , Cohort Studies , Cyclophosphamide/administration & dosage , Disease-Free Survival , Female , Humans , Immunological Synapses/immunology , Immunophenotyping , Leukemia, Prolymphocytic, T-Cell/blood , Leukemia, Prolymphocytic, T-Cell/genetics , Leukemia, Prolymphocytic, T-Cell/immunology , Male , Middle Aged , Piperidines/administration & dosage , Purines/administration & dosage , Quinazolinones/administration & dosage , Rituximab/administration & dosage , T-Lymphocytes/immunology , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
13.
Nat Commun ; 11(1): 3617, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32680998

ABSTRACT

Multiple myeloma (MM) progression is characterized by the seeding of cancer cells in different anatomic sites. To characterize this evolutionary process, we interrogated, by whole genome sequencing, 25 samples collected at autopsy from 4 patients with relapsed MM and an additional set of 125 whole exomes collected from 51 patients. Mutational signatures analysis showed how cytotoxic agents introduce hundreds of unique mutations in each surviving cancer cell, detectable by bulk sequencing only in cases of clonal expansion of a single cancer cell bearing the mutational signature. Thus, a unique, single-cell genomic barcode can link chemotherapy exposure to a discrete time window in a patient's life. We leveraged this concept to show that MM systemic seeding is accelerated at relapse and appears to be driven by the survival and subsequent expansion of a single myeloma cell following treatment with high-dose melphalan therapy and autologous stem cell transplant.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Clonal Evolution/drug effects , Hematopoietic Stem Cell Transplantation/adverse effects , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/pathology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Survival/drug effects , Cell Survival/genetics , Disease Progression , Dose-Response Relationship, Drug , Humans , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Middle Aged , Multiple Myeloma/diagnostic imaging , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Mutation/drug effects , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/therapy , Positron Emission Tomography Computed Tomography , Single-Cell Analysis , Spatio-Temporal Analysis , Transplantation, Autologous/adverse effects , Whole Genome Sequencing
14.
Cell Rep Med ; 1(1)2020 04 21.
Article in English | MEDLINE | ID: mdl-32483558

ABSTRACT

Clonal evolution of osimertinib-resistance mechanisms in EGFR mutant lung adenocarcinoma is poorly understood. Using multi-region whole-exome and RNA sequencing of prospectively collected pre- and post-osimertinib-resistant tumors, including at rapid autopsies, we identify a likely mechanism driving osimertinib resistance in all patients analyzed. The majority of patients acquire two or more resistance mechanisms either concurrently or in temporal sequence. Focal copy-number amplifications occur subclonally and are spatially and temporally separated from common resistance mutations such as EGFR C797S. MET amplification occurs in 66% (n = 6/9) of first-line osimertinib-treated patients, albeit spatially heterogeneous, often co-occurs with additional acquired focal copy-number amplifications and is associated with early progression. Noteworthy osimertinib-resistance mechanisms discovered include neuroendocrine differentiation without histologic transformation, PD-L1, KRAS amplification, and ESR1-AKAP12, MKRN1-BRAF fusions. The subclonal co-occurrence of acquired genomic alterations upon osimertinib resistance will likely require targeting multiple resistance mechanisms by combination therapies.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Carcinoma, Non-Small-Cell Lung , Clonal Evolution , Drug Resistance, Neoplasm/genetics , Lung Neoplasms , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Clonal Evolution/drug effects , Clonal Evolution/genetics , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , Female , Genetic Heterogeneity/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Protein Kinase Inhibitors/therapeutic use , Exome Sequencing , Young Adult
15.
Ann Hematol ; 99(7): 1551-1560, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32504186

ABSTRACT

TP53 aberrations reportedly predict favorable responses to decitabine (DAC) in acute myeloid leukemia (AML). We evaluated clinical features and outcomes associated with chromosome 17p loss or TP53 gene mutations in older, unfit DAC-treated AML patients in a phase II trial. Of 178 patients, 25 had loss of 17p in metaphase cytogenetics; 24 of these had a complex (CK+) and 21 a monosomal karyotype (MK+). In analyses in all patients and restricted to CK+ and MK+ patients, 17p loss tended to associate with higher rates of complete remission (CR), partial remission (PR), or antileukemic effect (ALE). Despite favorable response rates, there was no significant OS difference between patients with or without loss of 17p in the entire cohort or in the CK+ and MK+ cohort. TP53 mutations were identified in eight of 45 patients with material available. Five of the eight TP53-mutated patients had 17p loss. TP53-mutated patients had similar rates of CR/PR/ALE but shorter OS than those with TP53 wild type (P = 0.036). Moreover, patients with a subclone based on mutation data had shorter OS than those without (P = 0.05); only one patient with TP53-mutated AML had a subclone. In conclusion, 17p loss conferred a favorable impact on response rates, even among CK+ and MK+ patients that however could not be maintained. The effect of TP53 mutations appeared to be different; however, patient numbers were low. Future research needs to further dissect the impact of the various TP53 aberrations in HMA-based combination therapies. The limited duration of favorable responses to HMA treatment in adverse-risk genetics AML should prompt physicians to advance allografting for eligible patients in a timely fashion.


Subject(s)
Chromosome Deletion , Decitabine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Monosomy , Smith-Magenis Syndrome , Tumor Suppressor Protein p53/genetics , Aged , Aged, 80 and over , Chromosomes, Human, Pair 17/genetics , Clonal Evolution/drug effects , Clonal Evolution/genetics , DNA Mutational Analysis , Female , Germany/epidemiology , Humans , Karyotype , Karyotyping , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Monosomy/diagnosis , Monosomy/genetics , Mutation , Smith-Magenis Syndrome/diagnosis , Smith-Magenis Syndrome/epidemiology , Smith-Magenis Syndrome/genetics , Survival Analysis
16.
Blood ; 136(1): 36-49, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32430502

ABSTRACT

Acquired aplastic anemia (AA) and paroxysmal nocturnal hemoglobinuria (PNH) are pathogenically related nonmalignant bone marrow failure disorders linked to T-cell-mediated autoimmunity; they are associated with an increased risk of secondary myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Approximately 15% to 20% of AA patients and 2% to 6% of PNH patients go on to develop secondary MDS/AML by 10 years of follow-up. Factors determining an individual patient's risk of malignant transformation remain poorly defined. Recent studies identified nearly ubiquitous clonal hematopoiesis (CH) in AA patients. Similarly, CH with additional, non-PIGA, somatic alterations occurs in the majority of patients with PNH. Factors associated with progression to secondary MDS/AML include longer duration of disease, increased telomere attrition, presence of adverse prognostic mutations, and multiple mutations, particularly when occurring early in the disease course and at a high allelic burden. Here, we will review the prevalence and characteristics of somatic alterations in AA and PNH and will explore their prognostic significance and mechanisms of clonal selection. We will then discuss the available data on post-AA and post-PNH progression to secondary MDS/AML and provide practical guidance for approaching patients with PNH and AA who have CH.


Subject(s)
Anemia, Aplastic/pathology , Hemoglobinuria, Paroxysmal/pathology , Leukemia, Myeloid, Acute/etiology , Myelodysplastic Syndromes/etiology , Age of Onset , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Benzoates/adverse effects , Benzoates/therapeutic use , Bone Marrow/pathology , Chromosome Aberrations , Chromosomes, Human, Pair 7/genetics , Clonal Evolution/drug effects , Clone Cells/drug effects , Clone Cells/pathology , Disease Progression , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/genetics , Humans , Hydrazines/adverse effects , Hydrazines/therapeutic use , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Models, Biological , Monosomy , Mutation , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Oncogene Proteins, Fusion/genetics , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Selection, Genetic , Telomere Shortening
17.
Cancer Res ; 80(15): 3062-3069, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32245795

ABSTRACT

Next-generation sequencing has sparked the exploration of cancer genomes, with the aim of discovering the genetic etiology of the disease and proposing rationally designed therapeutic interventions. Driver gene alterations have been comprehensively charted, but the improvement of cancer patient management somewhat lags behind these basic breakthroughs. Recently, large-scale sequencing that focused on metastasis, the main cause of cancer-related deaths, has shed new light on the driving forces at work during disease progression, particularly in breast cancer. Despite a fairly stable pool of driver genetic alterations between early and late disease, a number of therapeutically targetable mutations have been found enriched in metastatic samples. The molecular processes fueling disease progression have been delineated in recent studies and the clonal composition of breast cancer samples can be examined in detail. Here we discuss how these findings may be combined to improve the diagnosis of breast cancer to better select patients at risk, and to identify targeted agents to treat advanced diseases and to design therapeutic strategies exploiting vulnerabilities of cancer cells rooted in their ability to evolve and drive disease progression.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , Clonal Evolution/genetics , Molecular Targeted Therapy , Mutation , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Clonal Evolution/drug effects , DNA Mutational Analysis , Disease Progression , Female , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Neoplasm Metastasis , Prognosis
19.
Cancer Res ; 80(7): 1578-1589, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31948939

ABSTRACT

A new ecologically inspired paradigm in cancer treatment known as "adaptive therapy" capitalizes on competitive interactions between drug-sensitive and drug-resistant subclones. The goal of adaptive therapy is to maintain a controllable stable tumor burden by allowing a significant population of treatment-sensitive cells to survive. These, in turn, suppress proliferation of the less-fit resistant populations. However, there remain several open challenges in designing adaptive therapies, particularly in extending these therapeutic concepts to multiple treatments. We present a cancer treatment case study (metastatic castrate-resistant prostate cancer) as a point of departure to illustrate three novel concepts to aid the design of multidrug adaptive therapies. First, frequency-dependent "cycles" of tumor evolution can trap tumor evolution in a periodic, controllable loop. Second, the availability and selection of treatments may limit the evolutionary "absorbing region" reachable by the tumor. Third, the velocity of evolution significantly influences the optimal timing of drug sequences. These three conceptual advances provide a path forward for multidrug adaptive therapy. SIGNIFICANCE: Driving tumor evolution into periodic, repeatable treatment cycles provides a path forward for multidrug adaptive therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Models, Biological , Neoplasms/drug therapy , Precision Medicine/methods , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Clonal Evolution/drug effects , Disease Models, Animal , Drug Administration Schedule , Drug Resistance, Neoplasm/genetics , Humans , Mice , Mutation Rate , Neoplasms/genetics
20.
Eur J Haematol ; 104(5): 488-498, 2020 May.
Article in English | MEDLINE | ID: mdl-31990086

ABSTRACT

INTRODUCTION: Therapy-related myelodysplastic syndrome and acute myeloid leukemia (t-MDS/AML) are defined as complications of previous cytotoxic therapy. Azacitidine (AZA), a hypomethylating agent, has showed activity in t-MDS/AML. OBJECTIVES: We evaluated the clonal dynamics of AZA-treated t-MDS/AML. METHODS: We collected bone marrow samples, at diagnosis and during treatment, from AZA-treated t-MDS/AML patients. NGS on 19 myeloid genes was performed, and candidate mutations with a variant allele frequency >5% were selected. RESULTS: Seven t-AML and 12 t-MDS were included with median age of 71 (56-82) years old, median number of AZA cycles of 6 (1-15), and median overall survival (OS) of 14 (3-29) months. We observed correlation between AZA response and clonal selection. Decrease of TP53-mutated clone was correlated with response to AZA, confirming AZA efficacy in this subgroup. In some patients, emergence of mutations was correlated with progression or relapse without impact on OS. Clones with mutations in genes for DNA methylation regulation frequently occurred with other mutations and remained stable during AZA treatment, independent of AZA response. CONCLUSION: We confirmed that the molecular complexity of t-MNs and that the follow-up of clonal selection during AZA treatment could be useful to define treatment combination.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Clonal Evolution/drug effects , Clonal Evolution/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Azacitidine/administration & dosage , Azacitidine/adverse effects , Biomarkers, Tumor , Female , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/diagnosis , Male , Mutation , Myelodysplastic Syndromes/diagnosis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL