scholarly journals Lenalidomide in non-Hodgkin lymphoma: biological perspectives and therapeutic opportunities

Blood ◽  
2015 ◽  
Vol 125 (16) ◽  
pp. 2471-2476 ◽  
Author(s):  
Athena Kritharis ◽  
Michael Coyle ◽  
Jaya Sharma ◽  
Andrew M. Evens

AbstractLenalidomide is an immunomodulatory drug (IMiD) with activity in lymphoid malignancies occurring primarily through immune modulation (eg, T-cell immune synapse enhancement and NK-cell/T-cell effector augmentation) and antiproliferative effects. Food and Drug Administration–approved for bortezomib-resistant, relapsed/refractory mantle-cell lymphoma, lenalidomide has demonstrated efficacy in several additional lymphoma subtypes. There are many ongoing clinical trials examining the use of lenalidomide alone or in combinatorial therapy. It will be important in these studies to delineate reliable, predictive biomarkers to optimally integrate lenalidomide into lymphoma treatment paradigms.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5299-5299
Author(s):  
Yonghong Zhang ◽  
Ling Jin ◽  
Jing Yang ◽  
Yanlong Duan ◽  
Chunjv Zhou ◽  
...  

Abstract One hundred and nineteen children with non-Hodgkin lymphoma were treated between February 2003 and December 2006 in Beijing Children’s Hospital on BCH-2003-NHL protocol. The diagnosis was made by histopathology of the biopsied tissue and/or bone marrow, and disease was classified according to WHO-2001 pathologic classification. We applied modified LMB89 protocol to cases with B-cell lymphoma; modified BFM90-ALL protocol for lymphoblastic lymphoma and cutaneous T-cell/NK cell lymphoma; and modified BFM90-ALCL protocol for anaplastic large-cell lymphoma (ALCL). There were 50 cases (42%) of B cell lymphoma including 32 cases of Burkitt¡’s lymphoma, 10 cases of Burkitt-like lymphoma and 8 cases of diffuse large B cell lymphoma; 44 cases (37%) of lymphoblastic lymphoma; 19 cases (16%) of ALCL; and 6 cases (5%) of cutaneous T-cell/NK cell lymphoma. The 85 boys and 34 girls (ratio, 2.5:1) ranged in age from 2 to 15 years (median, 7.8 years) at diagnosis. B cell lymphoma typically presented as abdomen mass and acute abdomen; nasopharynx and tonsil were also common sites of involvement. Lymphoblastic lymphoma generally presented with mediastinal mass and bone marrow involvement. There was no typical presentation for ALCL. According to the St. Jude staging system, 19 cases had stage I–II, and 94 cases stage III–VI diseases (exclude 6 cases of cutaneous T-cell/NK cell lymphoma). Seven cases had CNS involvement and 25 cases involved bone marrow. The treatment duration was 2 to 8 months for B-cell lymphoma, 2.5 to 3 years for lymphoblastic lymphoma and 1 to 1.5 years for ALCL. The follow-up rate was 100% and median observation period was 23 months. The overall survival (OS) at 3 years was 90.7% and the 3-year event-free survival (EFS) estimate was 82.3%. For B-cell lymphoma, 3-year OS was 88.68% and 3-year EFS was 81.8%. For lymphoblastoma lymphoma, the rates were 89.3% and 69.4%, respectively. All cases of ALCL are alive with on undergoing treatment for relapse. Patients with ALCL achieved the best 3-year OS (100%) and had 3-year EFS of 94.2%. Grade 3 or 4 bone marrow suppression occurred in 97.5% of patients with B-cell lymphoma, 100% of those with lymphoblastic lymphoma and 89.5% of cases with ALCL. As of to date, 11 patients have died, the causes of death include infection (n=4), abandonment of therapy (n=6) and relapse (n=1). Univarate analysis showed that stage IV disease, failure to achieve complete remission after 3 months of treatment, and bulky mass are were associated with poor prognosis £all P values <0.05£©. In summary, we have achieved excellent treatment results using modified international protocols. Infection and financial problem remained the main reasons of treatment failure.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 3043-3043
Author(s):  
Jianjun Gao ◽  
Hong Chen ◽  
Derek Ng Tang ◽  
Padmanee Sharma

3043 Background: Blockade of T cell co-inhibitory receptor CTLA-4 with a monoclonal antibody, ipilimumab, has led to augmented anti-tumor immune responses, clinical benefit, and FDA approval of ipilimumab for the treatment of metastatic melanoma. Only a subset of patients benefit from anti-CTLA-4 therapy. In order to identify genes, microRNAs, and signaling pathways that are modulated by anti-CTLA-4, which may be used for potential correlation with clinical outcomes or provide additional targets for therapy, we purified and analyzed CD4+T cells from patients treated with anti-CTLA-4 for changes in gene and microRNA expression profiles. Methods: On an IRB-approved phase Ia presurgical clinical trial, 6 patients with localized bladder cancer were treated with two doses of ipilimumab at 10 mg/kg at weeks 1 and 4. Pre-therapy and post-therapy blood samples were collected for CD4+ T cell enrichment by using the T cell isolation kit from Miltenyi Biotec (Auburn, CA). RNA and microRNA were isolated from purified CD4+T cells using Qiagen RNA isolation kits for Affymetrix microarray and micoRNA array analyses. Microarray data were then analyzed using Ingenuity iReport (Redwood City, CA). RT-PCR and Western blot were used to confirm significant changes in genes or pathways identified in microarray analyses. Results: Anti-CTLA-4 treatment resulted in modulation of differentially expressed genes (DEGs). After two doses of treatment, anti-CTLA-4 significantly changed expression of a total of 289 DEGs. Further pathway analyses indicated that anti-CTLA-4 induced a variety of pathways involved in cell proliferation and immune modulation, including PI3K/AKT, MAP/ERK, and IFN/JAK-STAT pathways. We have also identified 9 microRNAs that potentially regulate the expression of genes changed by anti-CTLA-4 therapy. Conclusions: Anti-CTLA-4 treatment results in modulation of multiple genes, microRNAs, and pathways, which likely play important roles in anti-tumor immune responses. We are currently testing a number of these identified genes and microRNAs as potential predictive biomarkers for anti-CTLA-4 therapy in a small cohort of patients who had complete response vs. progression of disease after anti-CTLA-4 therapy.


2013 ◽  
Vol 31 (6_suppl) ◽  
pp. 285-285
Author(s):  
Jianjun Gao ◽  
Hong Chen ◽  
Derek Ng Tang ◽  
Padmanee Sharma

285 Background: Blockade of T cell co-inhibitory receptor CTLA-4 with a monoclonal antibody, Ipilimumab (BMS), has led to augmented anti-tumor immune responses, clinical benefit, and FDA approval of Ipilimumab for the treatment of metastatic melanoma. Only a subset of patients benefit from anti-CTLA-4 therapy. In order to identify genes, microRNAs, and signaling pathways that are modulated by anti-CTLA-4, which may be used for potential correlation with clinical outcomes or provide additional targets for therapy, we purified and analyzed CD4+ T cells from patients treated with anti-CTLA-4 for changes in gene and microRNA expression profiles. Methods: On an IRB-approved Phase Ia presurgical clinical trial, 6 patients with localized bladder cancer were treated with two doses of Ipilimumab at 10 mg/kg at weeks 1 and 4. Pre-therapy and post-therapy blood samples were collected for CD4+ T cell enrichment by using the T cell isolation kit from Miltenyi Biotec (Auburn, CA). RNA and microRNA were isolated from purified CD4+ T cells using Qiagen RNA isolation kits for Affymetrix microarray and micoRNA array analyses. Microarray data were then analyzed using Ingenuity iReport (Redwood City, CA). RT-PCR and Western blot were used to confirm significant changes in genes or pathways identified in microarray analyses. Results: Ipilimumab treatment resulted in modulation of differentially expressed genes (DEGs). After two doses of treatment, Ipilimumab significantly changed expression of a total of 289 DEGs. Further pathway analyses indicated that Ipilimumab induced a variety of pathways involved in cell proliferation and immune modulation, including PI3K/AKT, MAP/ERK, and IFN/JAK-STAT pathways. We have also identified 9 microRNAs that potentially regulate the expression of genes changed by anti-CTLA-4 therapy. Conclusions: Ipilimumab treatment results in modulation of multiple genes, microRNAs, and pathways, which likely play important roles in anti-tumor immune responses. We are currently testing a number of these identified genes and microRNAs as potential predictive biomarkers for anti-CTLA-4 therapy in a small cohort of patients who had complete response vs. progression of disease after anti-CTLA-4 therapy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2619-2619
Author(s):  
Srividya Swaminathan ◽  
Line Dam Heftdal ◽  
Daniel F Liefwalker ◽  
Renumathy Dhanasekaran ◽  
Anja Deutzmann ◽  
...  

Abstract Background: The MYC oncogene drives T and B cell lymphoid malignancies, including Burkitt's lymphoma (BL) and Acute Lymphoblastic Leukemia (ALL). Such lymphomas are said to be "oncogene-addicted" to MYC. In order to develop targeted therapies for MYC-driven cancers, it is vital to understand how MYC regulates both cell autonomous and non-cell-autonomous processes, including host immunity. Approach: We have used a particularly tractable approach for studying the role of oncogenic MYC on the host immune system during lymphomagenesis through a tetracycline (tet)-system regulated transgenic mouse model of MYC-driven T cell lymphoma (SRα-tTA/Tet-O-MYC mice; Felsher and Bishop, Molecular Cell, 1999). By delineating the global immunological changes during primary MYC-driven T cell lymphomagenesis in SRα-tTA/Tet-O-MYC mice using mass cytometry (CyTOF) and CIBERSORT, we identified anti-tumor immune subsets that can be developed as therapies to treat MYC-driven lymphomas. Results: Amongst the immune subsets evaluated, our results demonstrated a specific systemic suppression of natural killer (NK) cell-mediated surveillance in SRα-tTA/Tet-O-MYC mice bearing overt MYC-driven T cell lymphomas. Inactivation of lymphoma-intrinsic MYC restores NK cell-mediated immune surveillance, suggesting that the regulation of NK cell-mediated surveillance may be integral to MYC-induced lymphomagenesis. We observed that lymphoma-specific MYC transcriptionally represses STAT1/2 and secretion of Type I Interferons (IFNs) required for normal NK cell homeostasis and NK cell-mediated immune surveillance in the tumor microenvironment. Concordantly, treating T cell lymphoma-bearing SRα-tTA/Tet-O-MYC mice with Type I IFN improves survival by rescuing NK cell production, and in part overriding MYC-mediated suppression of NK surveillance. We showed that human lymphomas with both high levels of MYC and low levels of STAT1/2 have lower NK surveillance, and are associated with poor prognosis. Finally, we established the therapeutic implications of our findings by showing that adoptive transfer of NK cells significantly delays lymphoma initiation, and recurrence after MYC inactivation; suggesting that NK cell-based therapy may be effective against MYC-driven lymphomas. Conclusion: Subversion of NK cell surveillance is integral to MYC-induced lymphomagenesis. Our studies provide a rationale for further developing the NK subset as a cell-based immunotherapy to effectively treat MYC-driven lymphomas in the future. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4539-4539
Author(s):  
Anne Valerie Decouvelaere ◽  
Franck Morschhauser ◽  
Marie-Christine Copin ◽  
Charles M. Dumontet

Abstract Protein Kinase C (PKC) is an important enzyme family. Twelve different isozymes have been described, which are implicated in diverse cell responses. Recently, studies have begun to define isoform-specific functions of PKC, and PKC beta II seems to play an antiapoptotic role. PKC beta II expression was analyzed by immunohistochemistry in 119 tumor samples of patients with lymphoid malignancies. The median age of the group was 55 years (range 14–85 y). Immunohistochemical staining was performed on formalin-fixed paraffin-embedded tissue, using the labeled avidin-biotin immuno-peroxidase complex method on the Ventana Benchmark automated immunostainer (Ventana Medical Systems, S.A., Illkirch, France). The sections were incubated with primary antibody (polyclonal antibody cPKC ßII (C-1): sc-210, Santa Cruz Biotechnology, Inc.) for 32 min at 1/100 dilution. The intensity of cytoplasmic and membrane immunostaining was assessed semi-quantitatively by two pathologists independently. Cases were considered “negative” when <10% of cells were stained, and “positive” when ≥10% of tumor cells showed cytoplasmic or membrane staining. Diffuse or heterogeneous staining was also specified. Intense and diffuse staining was observed in all mantle cell lymphomas (13 cases, 100%), and chronic lymphocytic lymphomas (CCL) (6 cases, 100%). PKC beta II was positive too in most follicular lymphomas (10 cases out of 11, 91%), although the pattern and the intensity of the staining were variable in accordance with the degree of lymphomatous infiltration. Indeed, in the same lymph node, the follicular lymphomatous areas were constantly labelled, whereas residual germinal centers remained negative. A majority of angioimmunoblastic T-cell lymphomas, lymphoblastic T-cell lymphomas and Marginal Zone/MALT lymphomas were labelled with anti-PKC beta II antibody, but the pattern of expression was more heterogeneous in these subtypes. A minority of diffuse large B-cell lymphomas expressed PKC beta II (8 out of 22 cases, including 4 with weak staining). Nearly all plasma cell neoplasms were negative. Nevertheless, 2 cases out of 16 (13%) displayed a diffuse moderate staining. In peripheral T-cell lymphomas, staining for PKC beta II was less frequent, as well as in primary cutaneous B and T lymphomas and pseudo-lymphomas. None of the cases of Hodgkin’s disease (13 cases) and anaplastic large cell lymphoma (5 cases) expressed PKC beta II. However, most of the numerous reactive cells were strongly stained. In conclusion, the intensity and the staining pattern of anti-PKC beta II antibody varied greatly according to the type lymphoid malignancy. These results highlight a significant correlation between PKC beta II expression and some particular type of lymphomas. In our series, the highest level of expression was found in mantle cell lymphomas and CLL.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3346-3346
Author(s):  
Scott J. Rodig ◽  
Jeremy S. Abramson ◽  
Geraldine S. Pinkus ◽  
Steven P. Treon ◽  
Margaret A. Shipp ◽  
...  

Abstract CD52 is a GPI-linked glycoprotein expressed by B cells, T cells, monocytes and macrophages. The humanized monoclonal antibody alemtuzumab (CAMPATH-1H) is specific for CD52 and is FDA-approved for the treatment of relapsed or refractory chronic lymphocytic leukemia (CLL). The utility of alemtuzumab in the treatment of other lymphoid and non-lymphoid malignancies has been recently explored; however, a comprehensive survey of CD52 expression among the various classes of hematopoietic neoplasms has not been completed. In addition, most methods of detecting CD52 rely on flow cytometric techniques that cannot be performed in a retrospective manner. Here we present methods for the reliable detection of CD52 on fixed, paraffin-embedded human bone marrow and lymphoid tissue samples using standard immunohistochemical staining methods and a non-humanized form of CAMPATH (CAMPATH-1G; Serotec, UK). Over 200 cases of B and T cell lymphomas and leukemias were evaluated. As summarized below (see table), the vast majority of low-grade B cell lymphoproliferative disorders (CLL/SLL, follicular lymphoma, lymphoplasmacytic lymphoma, mantle cell lymphoma) express CD52. Interestingly, some heterogeneity was noted within certain of these tumor types. For example, plasma cells were largely negative in lymphoplamsmacytic lymphomas, and proliferation centers were more intensely positive in CLL/SLL. In addition, we found that the majority of precursor B-cell acute lymphoblastic leukemia/lymphomas express this antigen. In contrast, there is some heterogeneity in CD52 expression among diffuse large B cell lymphomas, with approximately 22% of these tumors expressing undetectable or extremely low levels of CD52. Neoplasms of the T cell lineage also show marked heterogeneity in CD52 expression. The majority of precursor T cell acute leukemia/lymphomas and anaplastic large cell lymphomas showed no detectable CD52 expression; however, among the remaining T cell tumors, there was more pronounced tumor to tumor variation in CD52 expression. Among acute myeloid leukemias, we found CD52 expression only in AML-M4Eo. Finally, all cases of Hodgkin lymphoma and multiple myeloma tested were negative for CD52 expression. Together, these findings expand the subclasses of hematopoietic neoplasms that are rational candidates for alemtuzumab (CAMPATH-1H) therapy, and, in addition, reveal that CD52 is not universally expressed by lymphoid malignancies. Thus, target validation using this standard immunohistochemical procedure can now be readily performed on a case-by-case basis and is likely to be essential to guide alemtuzumab (CAMPATH-1H) therapy for hematological tumors. Expression of CD52 by Hematopoietic Neoplasms Diagnosis # Cases Tested # Cases CD52 Positive % Cases CD52 Positive * 2 of 2 Cases of AML-M4Eo positive CLL/SLL 25 25 100 Follicular Lymphoma 7 7 100 Mantle Cell Lymphoma 20 20 100 Lymphoplasmacytic Lymphoma 27 27 100 Burkitt Lymphoma 1 1 100 Diffuse Large B Cell Lymphoma 27 21 78 Pre-B ALL 18 16 89 Pre-T ALL 14 1 7 AML 23 2* 9 Peripheral T cell Lymphoma 18 7 39 Anaplastic Large Cell Lymphoma 6 0 0 Angioimmunoblastic Lymphoma 3 1 33 ATLL 2 2 100 Hepatosplenic T cell Lymphoma 1 0 0 NK/T Cell Lymphoma 2 1 50 Multiple Myeloma 18 0 0 Hodgkin Lymphoma 11 0 0


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4704-4704
Author(s):  
Qi Shen ◽  
Haitao Zheng ◽  
Shao hua Chen ◽  
Lijian Yang ◽  
Yangqiu Li

Abstract Abstract 4704 Elf-1 (E-74-like factor 1), a special transcription factor of T cells, is a member of the Ets (E-26-specific) family. Elf-1 plays a critical role in the transcription of the TCR ζ-chain. Our previous studies showed that the low expression level of TCR ζ-chain in hematologic malignancies, in the present study, we farther analyzed the expression of Elf-1 in patients with hematologic malignancies. Real-time PCR with SYBR Green I technique was used for detecting Elf-1 expression level in peripheral blood mononuclear cells from 33 patients with acute myeloid leukemia (AML) [including 17 cases with AML-M2 subtype (M2), 6 cases with (M3), 10 cases with (M5)], 49 patients with lymphoid malignancies [including T-cell acute lymphoblastic leukemia (T-ALL) 7 cases, T-cell non-Hodgkin lymphoma (T-NHL) 6 cases, B-cell acute lymphoblastic leukemia (B-ALL) 10 cases, B-cell chronic lymphoblastic leukemia (B-CLL) 7 cases, B-cell non-Hodgkin lymphoma (B-NHL) 19 cases] and 20 healthy volunteers. β2-microglobulin gene (β2M) was used as an endogenous reference. Relative changes in Elf-1 expression level were used by the 2-ΔCt×100% method. We found that the expression level of Elf-1 in 33 cases with AML (13.518±19.197%) was significant higher than those in the healthy control (2.044±1.321%) (P<0.01). However, the expression level of Elf-1 had no significant difference among the three AML subtypes (P>0.05). The expression level of Elf-1 in both T-cell lymphoid malignancies group (12.050±10.334%) and B-cell lymphoid malignancies group (14.144±21.576%) was significant higher than those in the healthy control (P<0.01, P<0.01). Moreover, the expression level of Elf-1 was different in AML and the lymphoid malignancies groups (P<0.01). That was, the highest in the B-ALL group, the lowest in the B-NHL group. These results indicated that overexpression of Elf-1 is a common feature in peripheral blood from patients with both AML and lymphoid malignancies, as TCR ζ-chain was down regulation in AML and lymphoid malignancies, the increased expression of Elf-1 gene, which an upstream factor of the TCRζ-chain might indicate a feedback up regulation situation in patients with hematologic malignancies. Disclosures: Shen: The study was supported by grants from National Natural Science Foundation of China (No. 30871091): Research Funding. Zheng:The study was supported by grants from National Natural Science Foundation of China (No. 30871091): Research Funding. Chen:The study was supported by grants from National Natural Science Foundation of China (No. 30871091): Research Funding. Yang:The study was supported by grants from National Natural Science Foundation of China (No. 30871091): Research Funding. Li:The study was supported by grants from National Natural Science Foundation of China (No. 30871091): Research Funding.


Digestion ◽  
2010 ◽  
Vol 81 (4) ◽  
pp. 231-234
Author(s):  
F. Mühr-Wilkenshoff ◽  
M. Friedrich ◽  
H.-D. Foss ◽  
M. Hummel ◽  
M. Zeitz ◽  
...  

2012 ◽  
Vol 2012 ◽  
pp. 1-9 ◽  
Author(s):  
Jessica M. McDaniel ◽  
Javier Pinilla-Ibarz ◽  
P. K. Epling-Burnette

The immunomodulatory agent, lenalidomide, is a structural analogue of thalidomide approved by the US Food and Drug Administration for the treatment of myelodysplastic syndrome (MDS) and multiple myeloma (MM). This agent is also currently under active investigation for the treatment of chronic lymphocytic leukemia (CLL) and non-Hodgkin’s lymphoma (NHL), as well as in drug combinations for some solid tumors and mantle cell lymphoma (MCL). Although treatment with lenalidomide has translated into a significant extension in overall survival in MM and MDS and has superior safety and efficacy relative to thalidomide, the mechanism of action as it relates to immune modulation remains elusive. Based on preclinical models and clinical trials, lenalidomide, as well as other structural thalidomide derivatives, enhances the proliferative and functional capacity of T-lymphocytes and amplifies costimulatory signaling pathways that activate effector responses and suppress inflammation. This paper summarizes our current understanding of T- and natural killer (NK) cell pathways that are modified by lenalidomide in hematopoietic neoplasms to inform future decisions about potential combination therapies.


Sign in / Sign up

Export Citation Format

Share Document