Checkpoint inhibitor therapy, with and without radiation, in diffuse large B cell lymphoma: A single-center analysis.

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e19059-e19059
Author(s):  
Stephanie Yoon ◽  
Mohammad Zahid ◽  
Pooja Phull ◽  
Jordan Senchak ◽  
Richard I. Fisher ◽  
...  

e19059 Background: Single agent checkpoint inhibitors (CPI) in NHLs have resulted in modest successes. Exceptions include RT, PCNS and testicular lymphomas, where higher activity is seen. As a single agent, CPI response rates range from 10-40% in relapsed refractory (RR) DLBCL, with few CRs and short response durations. In a cohort of RR DLBCL patients at our institution, we sought to identify clinical features that defined responders and non-responders. Methods: Between 9/2016 and 7/2018, 13 pts with DLBCL/RT, treated with a CPI, either on trial or as off-label therapy, with at minimum 1 infusion/cycle were included. Pathology specimens confirming DLBCL/RT were reviewed at FCCC. All pts had measurable disease by CT or PET/CT prior to CPI and had an evaluable response. Cell of origin was determined by Hans IHC. Results: Almost half (6/13) of pts achieved a response to CPI. Notably all responders had either concurrent or pre-treatment XRT. All 3 RT patients responded to CPI and continued to allo transplant. P3 and P6 both developed GVHD post allo, resulting in a demise in P3. No GC subtype pts responded nor had prior/ concurrent XRT with CPI. In 2 pts responses are ongoing, > 1 yr, and 1 RT pt remains in CR. Conclusions: XRT, prior or concurrent with CPI , was associated with durable responses in RR DLBCL. Patients with bulky ( > 7cm), rapidly progressive disease (8/13 cases) may require a 'debulking' strategy for CPI efficacy. An abscopal effect achieved with XRT/CPI combinations, may be impactful RR NHL. Analysis of PDL1/2 and MHC I/II, with other biomarkers, are underway. These clinical results warrant validation in a larger cohort, therefore a prospectively designed study is planned for 2019 in RR DLBCL/RT. [Table: see text]

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1720-1720
Author(s):  
Mayur Narkhede ◽  
Sadaf Qureshi ◽  
Maryam Yazdy ◽  
Roxanna Juarez ◽  
Giuseppe Esposito

Abstract Background DLBCL is the most common non-Hodgkin lymphoma (NHL), making up about 30%-40% of NHL in the U.S. PET-CT is recommended as the most accurate imaging technique in DLBCL for staging and response assessment. Pretreatment assessment of PET-CT scan derived metrics such as TMTV has been shown to correlate with PFS and/or overall survival (OS) in DLBCL (Sasanelli 2014) We attempted to replicate this finding using EFS at 24 months as a primary endpoint and compare it with pre-treatment TMTV, TLG and cell of origin (COO). Methods 47 pts with newly diagnosed DLBCL and treated with R-CHOP at our institution between 2014 to 2018 were identified from our electronic medical record system for retrospective analysis after IRB approval. All pts had a pretreatment PET-CT scan available for TMTV measurement. All pts had a pretreatment biopsy which were reviewed along with their clinical information regarding treatment outcome and follow up. Patients were classified as to germinal center B cell (GCB) and non-GCB based on immunochemistry using the Hahn's algorithm. PET-CT scans were reviewed by two nuclear medicine physicians using synovia software, and measurements for TMTV and TLG were recorded. TMTV was calculated using a threshold of 41% of the max pixel value (based on prior studies) to draw the volume of interest (VOI) for a lesion. Pooled t-test was performed to compare TMTV, TLG and COO with EFS at 24 mos. Chi-Square test compared TMTV with COO Results Median age of pts was 58 years, with a median duration of follow up of 26 months. There were 33% with limited stage (Stage I or II) and 67% were advanced stage (Stage III or IV). The mean pretreatment TMTV and pretreatment TLG was 295cm3 and 4519 units. 49% were GCB subtype and 47 % non-GCB. Amongst all patients 19.2 % had an event within 24 mos. When TMTV was compared to EFS at 24 months the mean TMTV was 304 for those who had an event versus 294 without (p=0.95). TLG compared to EFS at 24 months showed a mean TLG of 3391 for those who had an event versus 4914 without (P=0.40). GCB and non-GCB had mean TMTV of 264 and 339 respectively with p =0.59. COO when compared to TLG had means of 4365 and 4933 for GCB and non-GBB respectively with p=0.79.Whereas there was no correlation between stage and COO (p=0.4296) TMTV correlated with Ann Arbor staging (p=0.0002). Conclusion This retrospective study failed to demonstrate a correlation between pre-treatment TMTV, TLG, COO and EFS at 24 months revealing the lack of prognostic significance of pretreatment PET scan derived metrics in DLBCL. Prior studies with TMTV did not evaluate EFS at 24 months as an endpoint and therefore, longer follow up might be needed to demonstrate prognostic significance of pretreatment TMTV minimizing it clinical significance. The different subtypes of DLBCL based on COO as assessed by Hahns algorithm also did not differ in their disease burden as measured by TMTV. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 866-866 ◽  
Author(s):  
Jan A. Burger ◽  
Dan Landau ◽  
Julia Hoellenriegel ◽  
Carrie Sougnez ◽  
Matthias Schlesner ◽  
...  

Abstract The Bruton’s tyrosine kinase (BTK) inhibitor ibrutinib thwarts B cell receptor (BCR) signaling via irreversible inhibition of BTK, and induces durable remissions in relapsed/refractory CLL with a progression-free survival rate of 75% after 26 months of therapy (Byrd JC et al., NEJM 2013). However, a small fraction of patients treated with this targeted therapy develop progressive disease after an initial response. Here, we describe a longitudinal genomic investigation, utilizing whole-exome sequencing (WES) and copy number information of 3 patients who, with daily exposure to ibrutinib, achieved partial remission and later experienced CLL progression, but not Richter’s transformation. All 3 patients had advanced stage CLL (Rai stage 3-4) and were enrolled on IRB-approved phase 1/2 trials of ibrutinib (420 mg) as a single agent (Patients 1 and 2) or combined with rituximab (Patient 3). Patients 1 and 3 had relapsed diseased after prior FCR frontline therapy, while Patient 2 had received 4 prior lines of therapy. At treatment initiation, Patients 1 and 2 had known acquired TP53 deletions. Patient 1 additionally had del(13q), and Patient 2 a subclone (11.5% by FISH) carrying trisomy 12 and del(13q). Patient 3 had complex cytogenetics which included del(11q). As the best response to ibrutinib-based therapy, all three experienced partial responses. Patient 1 demonstrated normalization of hematologic parameters but experienced persistent bone marrow disease. Patient 2 achieved a >10-fold reduction but persistently elevated absolute lymphocyte count, and resolution of anemia and thrombocytopenia. Patient 3 had resolution of anemia, thrombocytopenia, and splenomegaly. Progressive disease was observed at days 1022, 554 and 206 following ibrutinib initiation for Patients 1-3, respectively. DNA was extracted from CD19-purified CLL cells before ibrutinib therapy and at the time of disease progression. Matched germline and tumor DNA from 2 timepoints underwent WES (mean coverage depth 170X) and copy number analysis (by SNP 6.0 arrays). Somatic alterations were identified through comparison with germline DNA. To examine clonal populations, we measured the allelic fractions of somatic variants and integrated this information with local copy number and purity information to infer the fraction of cancer cells (CCF) affected by the mutation. Since ibrutinib targets BTK, we searched for resistance-conferring mutations in the BTK gene in the progressing leukemias, such as the previously described C481S BTK mutation in 4 of 13 patients with acquired resistance (Chang et al., ASCO 2013, Abstract 7014). We observed that all three patients lacked mutations in BTK and for the most part, in other genes in the BCR signaling pathway. In Patient 2, we did identify a single nucleotide variant in PLCg2, a substrate of BTK previously reported to be mutated in a patient with ibrutinub resistance. However, the CCF affected by this mutation was smaller than 0.15, and therefore it is unlikely to be the main driver of relapse in this patient. All three CLLs acquired new somatic mutations at the time of progression not observed in the pre-treatment samples, involving recurrent lesions in CLL associated with poor clinical outcome. Patient 1 acquired a new clonal (>0.95 CCF) mutation in SF3B1 (K666T). Patients 2 and 3 revealed clonal deletions in chromosome 8p. Patient 2 additionally demonstrated an increase in a subclone harboring trisomy 12 with an associated MLL2 missense substitution, with CCF rising from 0.12 pre-treatment to 0.5 upon relapse. Our results confirm that clinically evident ibrutinib resistance cannot be uniformly attributed to mutations in BTK or other genes of the BCR signaling pathway. In the 3 CLLs presented herein, progressive disease was associated with the emergence of leukemic populations harboring genetic alterations with putative driver characteristics (del(8p), SF3B1 mutation) arising from a background of pre-existing 17p or 11q deletions. Our findings support the concept that CLL clones persisting during continuous therapeutic pressure can adapt to bypass BTK-related survival signaling. Ongoing studies focus on finer kinetic analysis of clonal dynamics in these patients during the period leading up to progressive disease to elucidate whether these alterations were newly acquired following ibrutinib exposure or represent selective expansions of pre-existing small subclones. Disclosures: Burger: Pharmacyclics: Research Funding. O'Brien:Pharmacyclics: Research Funding. Neuberg:Synta Pharmaceuticals: Trust owns stock; I am a Trustee Other.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 7514-7514 ◽  
Author(s):  
Kami J. Maddocks ◽  
Eva González Barca ◽  
Wojciech Jurczak ◽  
Anna Marina Liberati ◽  
Johannes Duell ◽  
...  

7514 Background: The Fc-enhanced CD19 antibody MOR208 and the immunomodulatory drug LEN have demonstrated single agent activity in patients (pts) with R-R DLBCL. MOR208 and LEN have shown synergy in vitro and in vivo in preclinical lymphoma models. This ongoing phase II study assesses the safety and efficacy of MOR208 + LEN in pts with R-R DLBCL. Methods: Pts >18 years old with R-R DLBCL, ECOG 0–2, adequate organ function, having previously received ≥1 but not more than 3 prior therapies, including ≥1 CD20-targeting regimen and who are not candidates for autologous stem cell transplant (ASCT), are eligible. Treatment comprises up to 12, 28-day (d) cycles (C) of MOR208 12 mg/kg IV, weekly during C1–3 (loading dose d4 of C1); every second week C4–12 + LEN 25 mg po d1–21, C1–12. Pts progression-free after 12 cycles receive up to 12 additional cycles of MOR208 (every second week). The primary endpoint is the overall response rate (ORR) by central radiology assessment. Secondary endpoints include disease control, duration of response, progression-free and overall survival, safety, and response by cell of origin and other biomarkers. A preplanned safety evaluation was undertaken. Results: 31 of 80 planned pts were enrolled prior to data cutoff (3 January 2017). Median age was 74 years (range 47–82); 45% of pts received ≥2 prior lines of therapy; 23% had rituximab refractory disease; 74% had Ann Arbor stage ≥III disease; 65% had elevated lactate dehydrogenase level, and 52% had a poor revised International Prognostic Index (3-5). The most common treatment-emergent adverse events (any grade/grade ≥3 [% pts]) were neutropenia (39/26), anemia (23/0) thrombocytopenia (16/6), infections (26/10) diarrhea (13/0), pyrexia (13/0), and rashes (13/6). Of 26 response evaluable pts (median follow-up 3.3 months), ORR (investigator assessed) was 58% (15 pts), with 7 (27%) complete responses. Median time to response was 1.8 months. Conclusions: The combination of MOR208 + LEN is well tolerated and shows promising activity in pts with R-R DLBCL. Accrual and follow-up of pts is ongoing, as are cell of origin and other biomarker analyses. Clinical trial information: NCT02399085.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 3112-3112
Author(s):  
Anthony B. El-Khoueiry ◽  
Nishan Tchekmedyian ◽  
Rachel E. Sanborn ◽  
Julio Antonio Peguero ◽  
Kerry Pulver ◽  
...  

3112 Background: Despite the broad activity of checkpoint inhibitors across tumor types, primary or secondary resistance after initial response represents a major challenge. Tomivosertib (T), a potent and highly selective inhibitor of the immunosuppressive kinases MNK-1 and 2, blocks expression of checkpoint proteins PD-1, PD-L1, and LAG-3 as well as immunosuppressive cytokines IL-6 and IL-8. In preclinical models, T was shown to trigger an anti-tumor immune response and enhance the activity of checkpoint inhibitors in a T-cell dependent manner. In prior clinical studies, T had an acceptable safety profile as a single agent and in combination with anti-PD-L1 agent avelumab. Methods: Patients experiencing insufficient response (progression or stable disease for 12 weeks or more) to any FDA-approved checkpoint inhibitor in any approved indication were eligible. T at 200 mg oral (PO) BID was added to the existing checkpoint inhibitor until disease progression or unacceptable toxicity was noted. Results: 39 pts (23 male, 16 female) were enrolled across seven cancer types. Median age was 68 (range 42-85). Median prior therapies were 2 (range 1-6). The most common cancers were lung (N = 17), urothelial (N = 6), renal (N = 5) and head and neck (N = 5). 36 pts continued on anti PD-1 antibody (Pembrolizumab and Nivolumab, 18 each) and 3 on anti PD-L-1 antibody (Durvalumab 2, Atezolizumab 1) . The most common grade 3/4 treatment related adverse events occurring in more than 1 pt were alanine aminotransferase increase (2), blood creatine phosphokinase increase (2) and maculo-papular rash (2). 7 patients discontinued treatment (18%) due to adverse events attributable to either drug. Three partial responses (PR) per RECIST 1.1 were observed in pts with previous progression on checkpoint inhibitor therapy, one each in NSCLC (1/17), gastric (1/1) and renal cancer (1/5). 7 NSCLC pts (41%) were progression free for ≥ 24 weeks. All NSCLC patients entered the study with progression by RECIST 1.1 on single agent checkpoint inhibitor prior to adding T. Conclusions: The addition of T to existing checkpoint therapy was well tolerated and manifested clinical activity including objective responses in pts with progression on existing checkpoint inhibitor. A Progression Free Survival rate at 24 weeks of 41% was noted in NSCLC patients. Additional studies evaluating the addition of T to checkpoint inhibitor therapy after progression on anti PD-1 or PD-L-1 therapy are planned. Clinical trial information: NCT03616834 .


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5637-5637
Author(s):  
Prakash Vishnu ◽  
Andrew Wingerson ◽  
David M Aboulafia

Abstract BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma (NHL). Recent advances in imaging, use of prognostic indices and molecular profiling has improved our ability to characterize disease and predict outcomes in DLBCL. About one-third of patients with DLBCL have bone marrow involvement at the time of diagnosis, and bone marrow aspirate/biopsy (BMAB) is considered gold standard to detect such involvement. 18 F-fluro-2-deoxy-D-glucose positron emission tomography combined with computed tomography (PET-CT), has become standard pre-treatment imaging in DLBCL and may be a non-invasive alternative to BMAB. Prior studies have suggested that PET-CT scan may obviate the need for BMAB as a component for staging patients with newly-diagnosed DLBCL, but owing to a variety of reasons this is not yet a standard of practice. We investigated whether FDG uptake-based bone marrow assessment can replace BMAB in newly-diagnosed DLBCL. METHODS This study is a single institution retrospective medical records' review. All patients with newly-diagnosed DLBCL at Virginia Mason Medical Center between January 2003 to December 2013 who underwent pre-treatment PET-CT and BMAB were included. FDG-PET/CT images were visually assessed for bone marrow involvement in posterior iliac crest. Patients with primary mediastinal DLBCL, previous history or coexistence of another lymphoma subtype and those with a non-diagnostic BMAB, and in whom the PET-CT did not show marrow signal abnormality were excluded from the analysis. Ann Arbor stage was determined using PET-CT with and without the contribution of BMAB, and the proportion of stage IV cases by each method was measured. RESULTS 105 eligible patients were identified. The median age was 62 years (range, 24-88), 62 (59%) were male, 53 (50%) had elevated LDH and 17 (16%) had an ECOG performance status of >2. Thirteen (12%) patients had > 1 extra-nodal site of lymphoma involvement. R-IPI score was 0-1 in 39 (37%), 2 in 42 (40%), 3 in 20 (19%), and 4 in 4 (4%) patients. A total of 38 (36%) patients had bone marrow involvement established by either PET-CT (n=24, 19%), BMAB (n=14, 13%), or both (n=12, 11%). 12 of the 24 patients (50%) with positive PET-CT had marrow involvement by DLBCL, while only 2 of the 81 patients (2%) with negative PET/CT showed marrow involvement. BMAB upstaged 1 of the 53 (2%) stage I/II patients to stage IV. The sensitivity of PET-CT scan to detect marrow involvement by DLBCL was 86% while the specificity was 87%. The positive predictive value of PET-CT was only 50% while the negative predictive value was 98%. CONCLUSIONS In patients with newly diagnosed DLBCL, PET-CT is complementary to BMAB in detecting marrow involvement by lymphoma. Although PET-CT has a high negative predictive value for bone marrow involvement, it overestimates the number of cases with marrow involvement by lymphoma. In clinical practice, routine BMAB may no longer be necessary for all patients with DLBCL, who are staged by PET-CT, unless the results would change both staging and therapy. The prognostic implication of marrow involvement identified by PET-CT compared to BMAB remains unknown. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Simone Camelliti ◽  
Valentino Le Noci ◽  
Francesca Bianchi ◽  
Claudia Moscheni ◽  
Francesca Arnaboldi ◽  
...  

Abstract Immune checkpoint inhibitors (ICIs) have made a breakthrough in the treatment of different types of tumors, leading to improvement in survival, even in patients with advanced cancers. Despite the good clinical results, a certain percentage of patients do not respond to this kind of immunotherapy. In addition, in a fraction of nonresponder patients, which can vary from 4 to 29% according to different studies, a paradoxical boost in tumor growth after ICI administration was observed: a completely unpredictable novel pattern of cancer progression defined as hyperprogressive disease. Since this clinical phenomenon has only been recently described, a universally accepted clinical definition is lacking, and major efforts have been made to uncover the biological bases underlying hyperprogressive disease. The lines of research pursued so far have focused their attention on the study of the immune tumor microenvironment or on the analysis of intrinsic genomic characteristics of cancer cells producing data that allowed us to formulate several hypotheses to explain this detrimental effect related to ICI therapy. The aim of this review is to summarize the most important works that, to date, provide important insights that are useful in understanding the mechanistic causes of hyperprogressive disease.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4443-4443 ◽  
Author(s):  
Seok Jin Kim ◽  
Hye Jin Kang ◽  
Dong-Yeop Shin ◽  
Dok Hyun Yoon ◽  
Kana Sakamoto ◽  
...  

Abstract Background: Hodgkin lymphoma (HL) and primary mediastinal large B-cell lymphoma (PMBCL) are curable disorder by systemic chemotherapy. However, 10-20% of patients experience treatment failure, and the treatment outcome of relapsed or refractory HL and PMBCL is still poor. Thus, the treatment of patients with relapsed and refractory disease remains challenging, and novel treatment strategies are needed. The Janus kinase-Signal transducer and activator of transcription (JAK/STAT) pathway has been suggested as a novel therapeutic target of these diseases because the activation of JAK/STAT and the in vitro efficacy of JAK2 inhibitor in HL and PMBCL were reported in previous pre-clinical studies. Given the importance of JAK/STAT signaling as a survival pathway in tumor cells, the use of JAK inhibitor may be a possible treatment option for relapsed or refractory patients with HL and PMBCL. Therefore, we performed a pilot study with ruxolitinib, JAK1/Jak2 inhibitor for relapsed or refractory HL and PMBCL. Methods: This study is a multi-center, single-agent, pilot study of ruxolitinib administered at a dose of 20 mg orally twice a day of a 28-day cycle. Patients with relapsed or refractory HL and PMBCL undergo treatment with ruxolitinib until disease progression or unacceptable toxicity are observed. Patients more than 18 years with measureable disease that was FDG-PET positive at baseline were eligible. Prior to the enrollment, patients should receive at least two types of previous treatment such as cytotoxic chemotherapy or autologous stem cell transplantation. Additional criteria included ECOG PS ≤2, adequate bone marrow, renal and hepatic function. The objectives of this study included characterization of anti-tumor activity and safety of ruxolitinib in relapsed or refractory HL and PMBCL patients. The response evaluation was performed according to Cheson criteria every two cycles, and the safety was assessed according to CTCAE v4. Details of the study are at ClinicalTrials.gov (NCT01965119). Results: To date, 10 patients (median age, 47 [range 21 - 70]) including 7 HL and 3 PMBCL have been treated with ruxolitinib between November 2013 and May 2014. Median number of prior therapies was 4 (2 - 6) and five patients relapsed after autologous stem cell transplantation. Prior to ruxolitinib treatment, all patients failed to show objective response to salvage treatment including progressive disease (n=7) and stable disease (n=3). All patients had baseline lymphadenopathy, and seven patients had extranodal involvement. Antitumor activity was observed in 3 of 7 patients with HL including three partial responses at the end of 2nd cycle scan. Among them, one patient exhibited progressive disease at the end of 4th cycle. However, the other two patients who maintained their response are continuing treatment at the time of analysis. Thus, they finished their 5th and 4thcycle, respectively, and updated results will be presented. The toxicity profile was manageable without any grade 3 or 4 hematologic and non-hematologic toxicity. Grade 1 toxicity included AST/ALT elevation (n=1), abdominal pain (n=1), myalgia (n=1) and sore throat (n=1). Grade 2 toxicity included neutropenia (n=1) and thrombocytopenia (n=2). The FISH study with tumor tissue of patients who participated in the study showed the amplified signals representing JAK1/JAK2 amplification. Conclusions: Ruxolitinib, a JAK1/JAK2 inhibitor, is well tolerated with an acceptable safety profile in patients with heavily treated HL and PMBCL, and shows significant single agent anti-tumor activity particularly for HL. Additional studies should be warranted to determine dosage and confirm efficacy of ruxolitinib in HL. Disclosures Off Label Use: Ruxolitinib (JAK2 inhibitor) for relapsed or refractory Hodgkin and mediastinal large B-cell lymphoma .


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 3138-3138
Author(s):  
Reem Abo-Zahrah ◽  
Daniel D. Karp ◽  
Abha Adat ◽  
Timothy A. Yap ◽  
Siqing Fu ◽  
...  

3138 Background: ERK1/2 signaling is often overactivated in cancer, especially in patients with molecular alterations activating the MAPK pathway. MAPK pathway inhibition can result in the increase of CD8+ and CD4+ T-cells and decreased expression of immunosuppressive cytokines. Methods: This is a retrospective study of 52 patients with advanced solid cancers and oncogenic alterations in the MAPK pathway, who were treated in phase I/II clinical trials with five different single agent ERK1/2 inhibitors at MD Anderson Cancer Center. We reviewed serial PET and/or CT imaging obtained before therapy, on therapy, and after therapy completion. We evaluated dynamic changes in the lymphatic nodes (LN) in the context of overall response per RECIST 1.1 and other outcomes. Results: Of the 52 patients, 19 (37%) patients were evaluated with serial PET/CT and 33 (63%) with serial CT imaging only. Of the 19 patients evaluated with PET/CT, 12 (63%) demonstrated increased FDG uptake in LN compared to pre-treatment imaging (LN enlargement, n = 9; no LN enlargement, n = 3) discrepant from the known target and non-target lesions. These 12 patients were on therapy with ERK inhibitors (11 at doses > recommended phase 2 dose [RP2D]) for a median of 3.6 months (range, 1.8-12 months) with a best response per RECIST 1.1. as follows: partial response, n = 1; stable disease (SD), n = 10; progressive disease (PD), n = 1. Of interest, in 6 of those 12 patients, FDG uptake in LN decreased or resolved after treatment discontinuation. Further, one patient had a biopsy of an emerged LN, which showed lymphocytic infiltrate without tumor cells. Of the 33 patients evaluated with CT only, 5 (15%) demonstrated increased size of LN discrepant from the known target and non-target lesions compared to pre-treatment imaging. These 5 patients were on therapy with ERK inhibitors (all at doses < RP2D) for a median of 1.4 months (range, 1.1-3.5 months) with a best response per RECIST 1.1. as follows: SD, n = 2; PD, n = 3. Of interest, in 2 of those 5 patients, size of LN decreased or resolved after treatment discontinuation. In addition, one patient had a biopsy of an emerged LN, which showed lymphoid aggerates without tumor cells. Conclusions: Our data suggest that treatment with ERK inhibitors can result in activation of the lymphatic nodes, which can manifest as pseudo-progression. This can lead to an inconclusive assessment of their therapeutic benefit and further suggests exploration of the potential synergistic effects with immune therapy.


2021 ◽  
Vol 14 (4) ◽  
pp. 288
Author(s):  
Laura Jimbu ◽  
Oana Mesaros ◽  
Cristian Popescu ◽  
Alexandra Neaga ◽  
Iulia Berceanu ◽  
...  

Checkpoint inhibitors were a major breakthrough in the field of oncology. In September 2014, based on the KEYNOTE-001 study, the Food and Drug Administration (FDA) approved pembrolizumab, a programmed cell death protein 1 (PD-1) inhibitor, for advanced or unresectable melanoma. Up until now, seven PD-1/PD-ligand(L)-1 inhibitors are approved in various solid cancers and hundreds of clinical studies are currently ongoing. In hematology, PD-1 inhibitors nivolumab and pembrolizumab were approved for the treatment of relapsed/refractory (R/R) classic Hodgkin lymphoma, and later pembrolizumab was approved for R/R primary mediastinal large B-cell lymphoma. In acute myeloid leukemia (AML), the combination of hypomethylating agents and PD-1/PD-L1 inhibitors has shown promising results, worth of further investigation, while other combinations or single agent therapy have disappointing results. On the other hand, rather than in first line, these therapies could be useful in the consolidation or maintenance setting, for achieving minimal residual disease negativity. Furthermore, an interesting application could be the use of PD-1/PD-L1 inhibitors in the post allogeneic hematopoietic stem cell transplantation relapse. There are several reasons why checkpoint inhibitors are not very effective in treating AML, including the characteristics of the disease (systemic, rapidly progressive, and high tumor burden disease), low mutational burden, and dysregulation of the immune system. We here review the results of PD-1/PD-L1 inhibition in AML and discuss their potential future in the management of this disease.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 392-392
Author(s):  
George Mulligan ◽  
Barb Bryant ◽  
Steven Roels ◽  
Andrew Bolt ◽  
Yan Meng ◽  
...  

Abstract Introduction: Clinical development of bortezomib in multiple myeloma (MM) included pharmacogenomic gene expression profiling (GEP) of pre-treatment tumor biopsies in order to evaluate the feasibility of developing predictive classifiers in prospective clinical trials and to better define the biological pathways related to drug sensitivity. By partitioning the samples into train (n=101) and test (n=80) datasets, statistically significant classifiers of response and survival were identified and then validated [Mulligan G, et al: Blood. 2007; 109:3177–88]. Here we describe specific genes and biological pathways associated with bortezomib efficacy in relapsed MM. Methods: Using tumor GEP data from 188 patients treated with single-agent bortezomib we identified genes associated with overall survival (OS), as well as genes associated with response (R) or progressive disease (PD) after bortezomib therapy. These clinical endpoints were also analyzed with biological pathway algorithms including GSEA [Subramanian A, et al: Proc Natl Acad Sci2005; 102:15545–50] Ingenuity Pathway Analysis and Gene Ontology/GATHER. These results were then compared to GEP data from a patient subset (n=76) treated with single-agent dexamethasone and also cell lines selected for differential sensitivity to bortezomib. Results: A comparison of R and PD highlighted numerous differentially expressed genes; approximately 200 were over-expressed in R and 500 were over-expressed in PD (t-test &lt;0.01). R-associated genes include signaling molecules like TRADD, CFLAR and TANK, while PD-associated genes included oncogenes (NRAS), apoptotic regulators (DAP3) and cancer antigens (CTAG). Pathway tools helped to integrate and extend these observations, consistently identifying NFkB activation and overexpression of adhesion pathways as R-associated. Overexpression of protein synthesis and mitochondrial pathways were PD-associated. The overexpression of pathways such as NFkB, adhesion and cytokines appear linked specifically with bortezomib and not dexamethasone sensitivity. These approaches were also applied to patient survival. In contrast to analyses of single genes, the pathway tools highlighted links between response and survival, including adhesion and inflammatory cytokine pathway expression. Conclusions: This study highlights the diversity of biological pathways expressed in MM and the utility of clinical genomics to associate pathways with activity of single-agent bortezomib.


Sign in / Sign up

Export Citation Format

Share Document