A Gene Expression-Based Risk Stratification Model Developed in Newly Diagnosed Multiple Myeloma Treated with High Dose Therapy Is Predictive of Outcome in Relapsed Disease Treated with Single Agent Bortezomib.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 656-656 ◽  
Author(s):  
Fenghaung Zhan ◽  
Barb Bryant ◽  
Bart Barlogie ◽  
George Mulligan ◽  
John D. Shaughnessy

Abstract Using data derived from the U133Plus2.0 microarray (U2), we recently constructed a 17-gene model predictive of high-risk multiple myeloma (MM). In the model, 13% of newly diagnosed cases were considered to have high-risk MM with 24 month overall survival estimates of 50% and 90% in the high- and low-risk groups, respectively (p<0.0001). Although validated in a separate cohort of newly diagnosed cases also receiving high dose therapy, it is not known whether this model has broader utility. To address this issue we applied the model to a dataset from a pharmacogenomic effort as part of a multi-center phase III trial testing the efficacy of the proteasome inhibitor bortezomib compared to dexamethasone. For purposes of evaluating the robustness of our model, this dataset is particularly useful in that it differs from the UAMS dataset in several important areas, such as relapsed vs. newly diagnosed disease, single agent bortezomib or dexamethasone vs. multi-agent induction therapy and high-dose melphalan-based tandem transplants, multiple centers vs. single center, delayed processing of bone marrow aspirates vs. immediate processing, negative vs. positive selection of plasma cells, and microarray platform U133A/B (UA) vs. U2 in the Millennium and UAMS datasets, respectively. A total of 144 of the first 351 UAMS cases applied to the U2 had also been applied to UA and were analyzed together with the 188 Millennium samples. Of the 17 genes on U2 we were able to find exact matches for 16 genes on UA. We applied the UA signal intensities of the 16 genes and the exact multivariate stepwise discriminant analysis (MSDA) model used to derive the 17-gene model using U2 data to the 144 UAMS cases. Risk scores for the 144 UAMS cases using the 17-gene U2- and 16-gene UA-derived models were calculated and a strong correlation observed (r=0.89; P<0.001). Using a score of greater than 1.6 as the high-risk cut-point with both models, a confusion matrix revealed a 96.5% agreement between high- and low-risk annotations among the 144 cases. Kaplan-Meier survival analysis of high- and low-risk groups defined by the 17- and 16-gene models revealed similar survival differences between high- and low-risk MM (U2: P=0.0046, HR=2.49; UA: P=0.0026; HR=2.58). These data suggests that the risk model is highly stable across these platforms. We then applied the 16-gene model to 188 cases of relapsed disease treated with bortezomib. Using the same discriminant score cut-off of 1.6 on the Millennium data, the 16-gene model defined 17.6% of cases as high-risk and this subset had significantly shorter overall survival times (P<0.0001; HR=2.93). The estimated 24-month survival was 9% in the high-risk vs. 40% in the low-risk cohort. The model also identified high-risk disease in a dexamethasone treated cohort (P<0.0001; HR=3.01). These data suggest that the 17-gene high-risk model is robust and not confounded by variables distinguishing these two datasets. These data also reveal a common molecular signature of high-risk MM in patients with newly diagnosed and relapsed disease pointing to a potential common molecular mechanism of drug resistance in MM that is revealed in this signature. We are currently developing and validating the 17-gene model using technology that quantifies tumor RNA directly in purified cells.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1499-1499 ◽  
Author(s):  
Catherine D. Williams ◽  
Jennifer L. Byrne ◽  
Gamal Sidra ◽  
Sonya Zaman ◽  
Nigel H. Russell

Abstract The established first-line treatment for younger myeloma patients includes regimens containing high-dose pulsed steroids and a combination of intravenous cytotoxic drugs such as vincristine and adriamycin (e.g.VAD). Response rates of 60% are reported but some patients are refractory. Patients may have complications relating to the need for central vascular access. Oral treatments, such as thalidomide, have shown response rates of about 36% in refractory patients. In an attempt to improve on these results we have evaluated the efficiency and toxicity of a novel oral chemotherapy regimen containing pulsed cyclophosphamide, thalidomide and pulsed dexamethasone (CTD). This regimen consists of a four week cycle of oral cyclophosphamide 500mg, given on days 1, 8 and 15, daily oral thalidomide initiated at a dose of 100mg daily for the first two weeks, increasing to 200mg daily if tolerated, and oral dexamethasone 40mg daily on days 1–4 and 15–18. Patients were treated with 2–6 cycles depending on response. Response criteria were used according to the EBMT/IBMTR guidelines. Results are reported on 62 myeloma patients: 15 with newly diagnosed (de novo) disease, 29 with VAD-refractory myeloma and 17 with relapsed disease. Median age of the patients was 55 years (range 31–73). Isotype was IgG in 38 patients, IgA in 17, light chain in 6 and non-secretory (NS) in 1 patient. Of the 15 patients with de novo disease, a median of 5 courses of CTD were given(range 4–6). All patients responded with 12 achieving a PR and 3 a CR. Of the 29 VAD-refractory patients, 14 had either no response or progressive disease after 2 cycles of VAD and 14 had only a minimal response to VAD with < 50% reduction in paraprotein after 3 cycles. One patient with NS myeloma had <50% reduction in bone marrow plasmacytosis after 3 cycles. PR or CR was achieved in 24 of the 29 patients (83%), with the remaining five achieving < 25% reduction in paraprotein. Twenty nine of the 44 de novo and refractory patients were considered suitable to progress to high dose melphalan and underwent stem cell mobilisation with cyclophosphamide and G-CSF. The median CD34+ cell dose harvested was 5.22 x 106/kg (1.9–11.2). Only one patient failed to mobilise. Twenty eight of these patients have undergone high dose therapy of whom 10 (36%) have achieved a CR. The relapsed disease group included 17 patients of whom 16 had received previous high-dose therapy. The overall response rate (PR/CR) was 71% (12/17). Ten patients went on to receive maintenance thalidomide and 2 a PSCT. Median follow-up is 19 months and 10 of the 17 are still alive. Toxicity of the CTD regimen was minimal with most adverse affects being grade 1 or 2 by the WHO classification. One third of patients suffered constipation and, a quarter, somnolence due to the thalidomide. Dose reduction was rarely required. Peripheral neuropathy was reported by 9% of patients.Grade 3 toxicity was limited to 2 patients who had a DVT and 2 patients who developed febrile neutropenia. Overall, CTD is a highly effective regimen for patients with newly diagnosed myeloma, VAD-refractory disease or relapsed disease. It is well tolerated and does not impair stem cell mobilisation. As it is oral, compliance is good and complications related to vascular access are minimised. This regimen has now gone on to be tested in new patients in a Phase III setting as one arm of the current MRC Myeloma IX trial.


2021 ◽  
Author(s):  
Hui Xiong ◽  
Weiting Kang ◽  
Qi Zhang

Abstract Background: This study aimed to explore N6-methyladenosine (m6A) methylation-related immune biomarkers and their clinical value in clear cell renal cell carcinoma (ccRCC).Methods: The RNA-seq data and clinical phenotype of ccRCC were downloaded from TCGA database. Immune-related genes list was downloaded from InnateDB database. Correlation analysis, survival analysis, univariate and multivariate Cox regression analysis were used to investigate the prognostic independent m6A-related immune genes, followed by prognosis risk model establishment. Patients were divided into high/low risk groups, followed by survival analysis, clinical factors, immune checkpoint genes and gene set variation analysis in high-risk vs. low-risk group. Results: Five prognostic independent m6A-related immune genes (PKHD1, IGF2BP3, RORA, FRK and MZF1) were identified. Low expression of PKHD1, RORA and FRK were associated with poor survival, while high expression of IGF2BP3 and MZF1 were associated with poor survival for ccRCC patients. Their expression showed correlations with multiple m6A genes. The risk model could stratify ccRCC patients into high/low risk group, and patients with high-risk were associated with short survival time. High-risk group had an high proportion of patients in tumor stage Ⅲ-Ⅳ and patients with pathologic T3-T4 tumors, lymph node metastasis (N1) and distant metastasis (M1). Ten immune checkpoint genes were differentially expressed in high/low risk groups, such as PD1 and CTLA-4. The risk group could be an independent prognostic factor (HR=1.69, 95% CI 1.07-2.68, P=0.0246). Conclusion: In this study, we developed a five genes risk model, which had independent prognostic value and associated with tumor stage, pathologic T/N/M and immune checkpoint expression in ccRCC.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 680-680 ◽  
Author(s):  
Shannon R. McCurdy ◽  
Jennifer A. Kanakry ◽  
Margaret M. Showel ◽  
Hua-Ling Tsai ◽  
Javier Bolaños-Meade ◽  
...  

Abstract Background: Recent advances in NMA haplo BMT, including high-dose PTCy for GVHD prophylaxis, have expanded potentially curative treatment options for patients (pts) without a matched donor. The DRI (Blood 2012;120:905), based on disease type and status, was developed to stratify pt risk across histologies and conditioning regimens. The current study evaluated the newly refined DRI (Blood 2014;123:3664) in the largest series of NMA haplo BMT to date. Methods: We retrospectively analyzed the outcomes of 374 consecutive adult hematologic malignancy pts who received NMA related haplo BMT with PTCy at Johns Hopkins. Eligibility included ECOG PS ≤ 2, LVEF ≥ 35%, adequate pulmonary and renal function and absence of uncontrolled infection. All pts received fludarabine (30 mg/m2IV D -6 to -2), Cy (14.5 mg/kg IV D -6 and -5), TBI (200 cGy D -1) and T-cell replete bone marrow. GVHD prophylaxis consisted of high-dose PTCy (50 mg/kg IV D 3 and 4), mycophenolate mofetil and tacrolimus. Maintenance therapy (e.g., imatinib) after engraftment was permitted. The median age was 55 (range 18-75) at BMT, 132 (35%) pts were aged ≥ 60 and 71 (19%) had prior autologous BMT. Diagnoses were acute leukemia or lymphoblastic lymphoma (115 pts; 31%), MDS or MPN (36 pts; 10%), aggressive NHL (95 pts; 25%), Hodgkin lymphoma (36 pts; 10%), mantle cell lymphoma (29 pts; 8%) and indolent lymphoid cancers (63 pts; 17%). By the new DRI, disease risk was low in 72 pts (19%), intermediate (int) in 242 (65%), high in 50 (13%) and very (v) high in 10 (3%). Overall results: With a median follow-up of 3.4 (range 0.5-11.4) years (y) in surviving pts, 3-y probabilities of PFS and OS were 40 (95% CI 35-45)% and 50 (45-56)%, respectively. The probability of neutrophil recovery was 89% by D30 (median 17 d). By competing-risk analyses, the D180 probability of nonrelapse mortality (NRM) was 8 (95% CI 5-10)%, grade 2-4 acute GVHD was 32 (27-37)%, and grade 3-4 acute GVHD was 4 (2-6)%. The 2-y probability of chronic GVHD was 13 (10-17)%. Results by DRI: Among the 3 DRI risk groups (low, int, high/v high), there were no statistically significant differences in histology (lymphoid vs myeloid), HCT-CI risk category, median CD34+ graft dose or pt CMV serostatus. Median pt age was greater in the higher risk groups (P = 0.01). On unadjusted analyses of the new DRI, the probability of relapse by competing-risk differed clearly between the groups (P < 0.0001; Fig. A), with no statistically significant difference in NRM (P = 0.53). This was coupled with statistically significant differences in both PFS and OS. In low, int and high/v high risk groups, 3-y PFS estimates were 64%, 37% and 22%, respectively (P < 0.0001; Fig. B), and 3-y OS estimates were 69%, 49% and 34% (P = 0.0001). Furthermore, on multivariate analyses adjusted for age and year of BMT, the new DRI was independently associated with overall outcomes. Compared to low-risk pts, the HR for relapse was 3.0 (95% CI 1.8-5.2) for int risk pts (P = 0.0001) and 4.7 (2.6-8.5) for high/ v high risk pts (P < 0.0001). HRs for both PFS and OS were ≥ 2-fold greater for int and high/v high risk pts compared to low risk pts (each P ≤ 0.0002). On multivariate analyses, the original DRI (13% low risk, 68% int, 19% high/v high) was also independently associated with relapse, PFS and OS (each P ≤ 0.003). When stratified by DRI, survival outcomes with reduced-intensity, matched related or unrelated BMT (based on 614 pts from the original DRI study cohort; personal communication, P. Armand) and haplo BMT with PTCy appeared similar (Table). Conclusion: The DRI appears to effectively stratify and prognosticate pts undergoing NMA haplo BMT. When stratified by DRI, the efficacy and survival outcomes with NMA haplo BMT with PTCy appear comparable to those reported with matched BMT. Within this transplant platform, the results support the use of the DRI in outcome analyses and randomized clinical trials involving heterogeneous groups of pts. Figure 1 Figure 1. Table Survival Estimates by DRI 3-y PFS (%) 3-y OS (%) Original DRI Matched Haplo Matched Haplo Low 66 63 70 71 Intermediate 31 39 47 49 High / very high 15 25 25 38 Figure 2 Figure 2. Disclosures Off Label Use: High-dose posttransplantation cyclophosphamide.


2021 ◽  
Vol 12 ◽  
Author(s):  
JingJing Zhang ◽  
Pengcheng He ◽  
Xiaoning Wang ◽  
Suhua Wei ◽  
Le Ma ◽  
...  

Background: RNA-binding proteins (RBPs) act as important regulators in the progression of tumors. However, their role in the tumorigenesis and prognostic assessment in multiple myeloma (MM), a B-cell hematological cancer, remains elusive. Thus, the current study was designed to explore a novel prognostic B-cell-specific RBP signature and the underlying molecular mechanisms.Methods: Data used in the current study were obtained from the Gene Expression Omnibus (GEO) database. Significantly upregulated RBPs in B cells were defined as B cell-specific RBPs. The biological functions of B-cell-specific RBPs were analyzed by the cluster Profiler package. Univariate and multivariate regressions were performed to identify robust prognostic B-cell specific RBP signatures, followed by the construction of the risk classification model. Gene set enrichment analysis (GSEA)-identified pathways were enriched in stratified groups. The microenvironment of the low- and high-risk groups was analyzed by single-sample GSEA (ssGSEA). Moreover, the correlations among the risk score and differentially expressed immune checkpoints or differentially distributed immune cells were calculated. The drug sensitivity of the low- and high-risk groups was assessed via Genomics of Drug Sensitivity in Cancer by the pRRophetic algorithm. In addition, we utilized a GEO dataset involving patients with MM receiving bortezomib therapy to estimate the treatment response between different groups.Results: A total of 56 B-cell-specific RBPs were identified, which were mainly enriched in ribonucleoprotein complex biogenesis and the ribosome pathway. ADAR, FASTKD1 and SNRPD3 were identified as prognostic B-cell specific RBP signatures in MM. The risk model was constructed based on ADAR, FASTKD1 and SNRPD3. Receiver operating characteristic (ROC) curves revealed the good predictive capacity of the risk model. A nomogram based on the risk score and other independent prognostic factors exhibited excellent performance in predicting the overall survival of MM patients. GSEA showed enrichment of the Notch signaling pathway and mRNA cis-splicing via spliceosomes in the high-risk group. Moreover, we found that the infiltration of diverse immune cell subtypes and the expression of CD274, CD276, CTLA4 and VTCN1 were significantly different between the two groups. In addition, the IC50 values of 11 drugs were higher in the low-risk group. Patients in the low-risk group exhibited a higher complete response rate to bortezomib therapy.Conclusion: Our study identified novel prognostic B-cell-specific RBP biomarkers in MM and constructed a unique risk model for predicting MM outcomes. Moreover, we explored the immune-related mechanisms of B cell-specific RBPs in regulating MM. Our findings could pave the way for developing novel therapeutic strategies to improve the prognosis of MM patients.


2021 ◽  
Author(s):  
Fang Wen ◽  
Xiaoxue Chen ◽  
Wenjie Huang ◽  
Shuai Ruan ◽  
Suping Gu ◽  
...  

Abstract Background: The diagnosis rate and mortality of gastric cancer (GC) are among the highest in the global, so it is of great significance to predict the survival time of GC patients. Ferroptosis and iron-metabolism make a critical impact on tumor development and are closely linked to the treatment of cancer and the prognosis of patients. However, the predictive value of the genes involved in ferroptosis and iron-metabolism in GC and their effects on immune microenvironment remain to be further clarified.Methods: In this study, the RNA sequence information and general clinical indicators of GC patients were acquired from the public databases. We first systematically screen out 134 DEGs and 13 PRGs related to ferroptosis and iron-metabolism. Then, we identified six PRDEGs (GLS2, MTF1, SLC1A5, SP1, NOX4, and ZFP36) based on the LASSO-penalized Cox regression analysis. The 6-gene prognostic risk model was established in the TCGA cohort and the GC patients were separated into the high- and the low-risk groups through the risk score median value. GEO cohort was used for verification. The expression of PRDEGs was verified by quantitative QPCR.Results: Our study demonstrated that patients in the low-risk group had a higher survival probability compared with those in high-risk group. In addition, univariate and multivariate Cox regression analyses confirmed that the risk score was an independent prediction parameter. The ROC curve analysis and nomogram manifested that the risk model had the high predictive ability and was more sensitive than general clinical features. Furthermore, compared with the high-risk group, the low-risk group had higher TMB and a longer 5-year survival period. In the immune microenvironment of GC, there were also differences in immune function and highly infiltrated immune cells between the two risk groups.Conclusions: The prognostic risk model based on the six genes associated with ferroptosis and iron-metabolism has a good performance for predicting the prognosis of patients with GC. The treatment of cancer by inducing tumor ferroptosis or mediating tumor iron-metabolism, especially combined with immunotherapy, provides a new possibility for individualized treatment of GC patients.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3394-3394
Author(s):  
Wei Xiong ◽  
Fenghuang Zhan ◽  
Yongsheng Huang ◽  
Bart Barlogie ◽  
John D. Shaughnessy

Abstract Deletion of chromosome 17p13 is a poor risk feature in MM, presumably resulting from a loss-of-hetrozygosity of the TP53 locus. However, details on the role of TP53 in the pathogenesis of MM and its relevance as a prognostic variable remain uncertain. We determined TP53 mRNA expression, DNA sequence integrity and copy number in newly diagnosed and relapsed disease and correlated these features with disease progression and outcome. Among 351 newly diagnosed cases treated on TT2, a randomized trial of high-dose chemotherapy and autotransplants +/− thalidomide. TP53 gene expression, as determined by Affymetrix microarray of CD138-selected cells, varied from a high of 5,241 to a low of 10 (mean 1,460). TP53 expression lower than 727, present in 10% of tumors, was associated with shorter event-free survival (EFS: P=0.0012) and overall survival (OS: P=0.0006) with a median follow-up of 40 months. Short EFS (P<0.0538) and OS (P<0.0384) was also associated with a low TP53 expression (< 727) in 10% of 214 cases treated on TT3 followed a median of 16 months. On univariate analyses, low TP53 was linked to high CRP (>4.1 mg/L), creatinine (>221 umol/L), and LDH (>191 IU/L), Amp1q21, del13, MRI lesions (>3), and high-risk molecular groups (PR, MS, and MF) (P < .05) in the TT2 cohort. There was no significant difference in cytogenetic abnormalities, B2M (>4 mg/L), albumin (<3.5 g/dl), or marrow plasmacytosis (>33%). On multivariate regression analysis, low TP53 expression was an independent variable predicting shorter survival (EFS: P=0.044; OS: P=0.019), even in the context of high-risk molecular entities and ISS. We next used interphase FISH to correlate TP53 gene copy number with expression in randomly selected cases from the TT2 cohort. 17p13 deletion was observed in 88% (27% with bi-allelic deletion) of 17 cases from the group of 35 (10% of total) with the lowest TP53 expression, while deletion was present in 36% in 22 cases with low-, 12% in 17 cases with mid-level, and 6% in 17 samples with high-expression of TP53 (P<0.001). Surprisingly, there was no correlation between TP53 gene expression levels and expression of 121 known TP53 targets genes in newly diagnosed and relapsed disease, yet a strong correlation was observed in myeloma cell lines. Sequence analysis of TP53 revealed no mutations in 24 newly diagnosed cases, whereas mutations were seen in exon 7, 8 and 9 in 5 of 20 relapsed samples (P=0.014). Analysis of TP53 expression in 90 relapses from the TT2 cohort revealed that 18% had TP53 at or below the high-risk level of 727, representing a near doubling of the percentage seen in newly diagnosed disease (P=0.019). Comparison of TP53 expression in 50 paired baseline-relapse samples from the TT2 cohort revealed a near significant trend for a reduction across 32 relapses from the no thalidomide arm (P=0.08) and a significant increase in expression across 18 relapses on the thalidomide arm (P=0.0035). Sequence analysis of TP53 in seven paired baseline and relapse samples from cases treated with thalidomide revealed no mutations were observed. A more comprehensive sequence and copy number analysis on paired samples from both arms is pending. Together these data demonstrate that low TP53 expression is strongly correlated with 17p13 deletion and is a significant and independent prognostic variable, independent of changes in TP53 target genes.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1312-1312 ◽  
Author(s):  
Gary H. Lyman ◽  
Jeffrey Crawford ◽  
Nicole M. Kuderer ◽  
Debra Wolff ◽  
Eva Culakova ◽  
...  

Abstract Introduction: Neutropenic complications including severe neutropenia (SN) and febrile neutropenia (FN) represent major dose-limiting toxicities of cancer chemotherapy. A prospective study was undertaken to develop and validate a predictive model for neutropenic events in patients receiving cancer chemotherapy. The final risk model based on mature data is presented. Methods: Between 2002 and 2006, 4458 consenting patients starting a new chemotherapy regimen at 115 randomly selected community oncology practices throughout the United States were enrolled including 3760 with cancers of breast, lung, colorectum, ovary and malignant lymphoma receiving at least one cycle of treatment. Using a 2:1 random split sample methodlogy, a risk model for first-cycle SN or FN was derived and validated based on multivariate logistic regression analysis incorporating pretreatment variable information. The cumulative risk of events over the initial 120 days of treatment was estimated by the method of Kaplan and Meier. High and low risk groups were defined on the basis of the median predicted risk and model test performance characteristics were estimated. Results: Following adjustment for cancer type, important predictive factors included: older age, prior chemotherapy, abnormal hepatic or renal function, low pretreatment white blood count, immunosuppressive medications and planned relative dose intensity &gt;85% as well as use of several specific chemotherapeutic agents including anthracyclines, taxanes, alkylating agents, topoisomerase inhibitors, gemcitabine or vinorelbine. Lower risk of neutropenic complications were associated with primary prophylaxis with a colony-stimulating factor (CSF). Individual risk estimates based on the model ranged from 0–89% with mean and median of 19.2% and 10.1%, respectively. The model was associated with an R2 of 0.34 and demonstrated excellent discrimination with a c-statistic of 0.833 [95% CI: 0.813–0.852, P&lt;.001]. The model predicted risk of cycle 1 SN or FN in high and low risk groups was of 34% and 4%, respectively. The cumulative risk of FN over the initial 120 days was 20% in high risk patients and 5% in low risk patients. Model performance included sensitivity and specificity of 90% and 59%, respectively, with a model diagnostic odds ratio of 12.8 [95% CI: 9.3, 17.7]. Application of the model to the validation data set was associated with similar excellent discrimination and test performance characteristics. CSF prophylaxis applied to high risk patients was associated with significantly lower risk of FN over repeated cycles of chemotherapy [HR = 0.51; 95% CI: 0.35 – 0.75; P &lt;.0001]. Nearly two-thirds of patients classified as high risk but who did not receive primary CSF prophylaxis went on to receive secondary use during subsequent cycles. Discussion: Based on excellent test performance characteristics, the risk model identified patients with a cumulative incidence of FN of at least 20% who are candidates for targeted prophylaxis with a CSF. Further validation of this model in actual clinical practice is currently underway.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 733-733
Author(s):  
John D. Shaughnessy ◽  
Pingping Qu ◽  
Ricky Edmondson ◽  
Damir Herman ◽  
Yiming Zhou ◽  
...  

Abstract Introduction: The mechanism of action and resistance to chemotherapy is poorly understood and measures of efficacy typically rely on clinical outcome data. Recent advances suggest that prospective gene expression profiling (GEP) can be used to more accurately define not only short-term but lasting treatment benefits. We recently reported that baseline tumor cell gene signatures, encompassing 70 and as few as 17 genes, can discriminate risk groups of myeloma patients both in the untreated and previously treated settings. However, a subset of predicted low-risk cases followed an aggressive clinical course accompanied by a shift from 70-gene-defined low-to high-risk over time, either reflecting clonal evolution or outgrowth of aggressive clones present, but undetectable, at diagnosis. Accurately identifying this patient population is a first step in preempting such transformation. We hypothesized that changes in gene expression patterns of tumor cells following a short term in-vivo challenge with a single agent chemotherapeutic might expose these latently aggressive cells. Unlike in-vitro testing, clinical drug administration also allows for assessing tumor cell perturbation in the context of host interactions. Building on recent observations that clinical outcome in myeloma patients could be correlated with 48hr GEP changes induced in vivo following single agent administration of thalidomide, lenalidomide, and dexamethasone, we now examined whether such short-term tumor-cell GEP and proteomic alterations could fine-tune clinical outcome prediction beyond the well established 70-gene-based baseline prediction model. Methods: Affymetrix U133Plus2.0 microarray analysis and mass spectrometry were performed on CD138-enriched tumor cells prior to and 48hr following a single test-dose application of bortezomib at 1.0mg/m2 in 142 newly diagnosed patients with MM. A total of 1051 genes (P < .005) were differentially expressed at 48 hours. Both change in expression, adjusting for baseline expression, and post-drug expression levels of each gene were examined for correlations with event-free survival in a Cox proportional hazards model. Post-drug expression was chosen and 113 genes were retained (p <= .05). The difference of the mean log2 expression of genes with hazard ratios (HR) of < 1.0 (favorable) and genes with HR >=1 (unfavorable) was used to create a score which, in the context of running log rank statistics, was used to classify patients into high- and low-risk groups. The independent prognostic power of the score for event-free and overall survival was investigated, together with baseline prognostic variables, by multivariate analysis. This method was tested in a 10-fold cross-validation procedure using the same data set. The model is currently being validated in an independent set of 100 cases and results will be reported. Results: Changes in the expression of proteasome genes, and their related proteins, predominated a list of 113 outcome-related genes. A high-risk score, associated with upregulation of proteasome genes, seen in 24% of cases, was associated with median survival of less than 24 months, dramatically contrasting with a 3-year survival estimate of greater than 80% in the 76% in whom proteasome genes were not activated (p<0.0001). The cross-validated post-bortezomib score was an independent predictor of outcome in multivariate analysis of standard and genetic variables, including the well established and validated baseline 70-gene risk score. Importantly, 12% of patients in the 70-gene model-defined low-risk category were upgraded to high-risk by the 113-gene post-bortezomib model, with poor outcomes resembling those in the 70-gene-defined high-risk baseline model. Moreover, the 113-gene post-bortezomib score alone accounted for an unprecedented 57% of outcome variability by R2 statistics, with hazard ratio values of 5.45 for overall and 7.84 for event-free survival. Conclusions: The rapid activation of proteasome genes and their corresponding proteins in MM cells within 48hr of a single bortezomib test-dose exposure as an indicator of poor clinical outcome suggests a novel and perhaps central mechanism of resistance to this new class of cancer therapeutics and perhaps standard genotoxic agents, which were part of the overall treatment. We are now testing the hypothesis whether the high-risk associated with the post-bortezomib proteasome activation can be overcome by higher doses of bortezomib or the addition of agents targeting other critical pathways.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4497-4497
Author(s):  
Luis Antonio Meillon ◽  
Salvador Campos-Cabrera ◽  
Nancy Delgado-Lopez ◽  
Angel A. Fernandez-Martinez ◽  
Jesus Medrano-Contreras ◽  
...  

Abstract Abstract 4497 Background: IM 400mg daily is the current standard of treatment of CP CML. Almost 70% of patients achieve complete cytogenetic response (CCR) at 12 months (mo) (IRIS Trial) and the rate of Major Molecular Response (MMR) is 40% with IM 400mg daily (TOPS Trial). On the other hand, low doses of Peg-IFN 2a has the same efficacy and lower toxicity profile and cost as high dose, mainly when it is combined with other drugs (MRC and Hovon groups, Blood 2004; 103:4408). Combination of IM + Peg-IFN 2a or Cytarabine could increase the rate of response in CML. Aims: To evaluate the rates of CCR and MMR at 12 mo in Mexican patients with newly-diagnosed CP-CML treated with IM versus IM + Peg-IFN 2a. Methods: Between 2006 and 2008, 10 patients were randomized 1:1 and stratified by Sokal and Hasford risk groups. Cytogenetic and Molecular assesments were available for all patients. In the arm A of the study IM 400 mg daily was delivered. In the Arm B IM + Peg-IFN 2a was delivered at 400 mg daily + 90 μ g per week respectively. Peg-IFN 2a was started in the 7th week of treatment (Hochhaus A, et al ASCO 2003. Abstract #2287). Results: For this initial report, 10 patients were analyzed, median age was 35 years (25-55), 50% males; Sokal score was: low risk 70%, and high risk 30%. Hasford score was: low risk 40%, intermediate 30% and high risk 30%. All patients are alive with a median follow-up of 41 mo (30-48). CCR at 12 mo was obtained in 80% in both arms of treatment. MMR was obtained in 10% in patients with IM alone and 30% in patients with IM + Peg-IFN 2a. Overall the rate of primary resistance was 10% and the rate of suboptimal response was 10%. IM 400 mg was well tolerated. Peg-IFN 2a was tolerated in most patients, but needed discontinuation because increased cytopenias or other non hematologic toxicities. The median time of duration of treatment with Peg-IFN 2a was 6 mo (3-12). Conclusions: IM 400 mg and IM 400 mg + Peg-IFN 2a obtained similar rates of CCR. IM 400 mg + Peg-IFN 2a obtained a higher rate of MMR. The combination of IM 400 mg + Peg-IFN 2a is feasible. There is a slight increase in toxicity with this combination. Further follow-up is warranted for these patients. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document