{"title":"Defective Immunity Against SARS-CoV-2 Omicron Variants Despite Full Vaccination in Hematologic Malignancies.","authors":"Laurence Zitvogel, Lisa Derosa, Guido Kroemer","doi":"10.1158/2643-3230.BCD-22-0213","DOIUrl":"10.1158/2643-3230.BCD-22-0213","url":null,"abstract":"<p><strong>Summary: </strong>In patients with multiple myeloma, completion of mRNA-based vaccination schemes failed to yield detectable SARS-CoV-2 Omicron-neutralizing antibodies and S1-RBD-specific CD8+ T cells in approximately 60% and 80% of the cases, respectively. Patients who develop breakthrough infections exhibited very low levels of live-virus neutralizing antibodies and the absence of follicular T helper cells. See related article by Azeem et al., p. 106 (9). See related article by Chang et al., p. 1684 (10).</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":"4 3","pages":"172-175"},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10150292/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9875791","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Francesco Maura, Bachisio Ziccheddu, Jenny Z Xiang, Bhavneet Bhinder, Joel Rosiene, Federico Abascal, Kylee H Maclachlan, Kenneth Wha Eng, Manik Uppal, Feng He, Wei Zhang, Qi Gao, Venkata D Yellapantula, Vicenta Trujillo-Alonso, Sunita I Park, Matthew J Oberley, Elizabeth Ruckdeschel, Megan S Lim, Gerald B Wertheim, Matthew J Barth, Terzah M Horton, Andriy Derkach, Alexandra E Kovach, Christopher J Forlenza, Yanming Zhang, Ola Landgren, Craig H Moskowitz, Ethel Cesarman, Marcin Imielinski, Olivier Elemento, Mikhail Roshal, Lisa Giulino-Roth
{"title":"Molecular Evolution of Classic Hodgkin Lymphoma Revealed Through Whole-Genome Sequencing of Hodgkin and Reed Sternberg Cells.","authors":"Francesco Maura, Bachisio Ziccheddu, Jenny Z Xiang, Bhavneet Bhinder, Joel Rosiene, Federico Abascal, Kylee H Maclachlan, Kenneth Wha Eng, Manik Uppal, Feng He, Wei Zhang, Qi Gao, Venkata D Yellapantula, Vicenta Trujillo-Alonso, Sunita I Park, Matthew J Oberley, Elizabeth Ruckdeschel, Megan S Lim, Gerald B Wertheim, Matthew J Barth, Terzah M Horton, Andriy Derkach, Alexandra E Kovach, Christopher J Forlenza, Yanming Zhang, Ola Landgren, Craig H Moskowitz, Ethel Cesarman, Marcin Imielinski, Olivier Elemento, Mikhail Roshal, Lisa Giulino-Roth","doi":"10.1158/2643-3230.BCD-22-0128","DOIUrl":"10.1158/2643-3230.BCD-22-0128","url":null,"abstract":"<p><p>The rarity of malignant Hodgkin and Reed Sternberg (HRS) cells in classic Hodgkin lymphoma (cHL) limits the ability to study the genomics of cHL. To circumvent this, our group has previously optimized fluorescence-activated cell sorting to purify HRS cells. Using this approach, we now report the whole-genome sequencing landscape of HRS cells and reconstruct the chronology and likely etiology of pathogenic events leading to cHL. We identified alterations in driver genes not previously described in cHL, APOBEC mutational activity, and the presence of complex structural variants including chromothripsis. We found that high ploidy in cHL is often acquired through multiple, independent chromosomal gains events including whole-genome duplication. Evolutionary timing analyses revealed that structural variants enriched for RAG motifs, driver mutations in B2M, BCL7A, GNA13, and PTPN1, and the onset of AID-driven mutagenesis usually preceded large chromosomal gains. This study provides a temporal reconstruction of cHL pathogenesis.</p><p><strong>Significance: </strong>Previous studies in cHL were limited to coding sequences and therefore not able to comprehensively decipher the tumor complexity. Here, leveraging cHL whole-genome characterization, we identify driver events and reconstruct the tumor evolution, finding that structural variants, driver mutations, and AID mutagenesis precede chromosomal gains. This article is highlighted in the In This Issue feature, p. 171.</p>","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":"4 3","pages":"208-227"},"PeriodicalIF":11.5,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10150291/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9888501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kecheng Zhang, M. Rees, M. Khanlari, Laura Rooms, Raul C. Ribeiro, M. Wlodarski
{"title":"Abstract A38: Pediatric Germline CSF3R T618I mutation gives insight into disease progression and long-term therapy options of Chronic Neutrophilic Leukemia","authors":"Kecheng Zhang, M. Rees, M. Khanlari, Laura Rooms, Raul C. Ribeiro, M. Wlodarski","doi":"10.1158/2643-3249.aml23-a38","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a38","url":null,"abstract":"\u0000 Chronic Neutrophilic Leukemia (CNL) is a rare BCR-ABL negative myeloproliferative neoplasm that affects approximately 1 in a million individuals. The typical age of onset and disease presentation is in elderly patients with a median age of 63 and a range of 9-84. The oncogenic driver of CSF3R, the G-CSF receptor protein, in CNL has been classified over the last decade and has improved therapies for patients with targeted Tyrosine Kinase Inhibitors, such as Ruxolitinib (a potent JAK1/2 inhibitor). Currently, the only curative option is an allogeneic hemopoietic stem cell transplant with typical median overall survival of 2 years. Because of the low occurrence rate of CNL, there have only been 3 germline cases of CSF3R classified in literature. With these patients, they exhibit a lifelong skewing of the myeloid lineage causing gross neutrophilia. Our patient presented at 13 months with leukocytosis and neutrophilia, resulting in genetic testing revealing the diagnosis of the youngest documented case of CNL. A germline CSF3R T618I constitutive activating mutation was documented in our patient and in her father. Her father had documented lifelong leukocytosis of indeterminate origin and it was not until 33 that genetic testing was done, and he was diagnosed with CNL. He underwent an allogeneic stem cell transplant after being admitted with gross hepatosplenomegaly with a WBC of 322, the highest recorded in a germline CNL patient in literature. Our patient presented to clinic with leukocytosis ranging from 45-98 and hepatosplenomegaly. After a bone marrow aspirate and biopsy was taken to determine disease stage and severity, we immediately began Ruxolitinib therapy. We began 20 mg BID dosing based on platelets and monitored response to therapy while conducting targeted RNA-seq for myelodysplastic and myeloid leukemia genes on the bone marrow, which did not detect any secondary mutations. Ruxolitinib dosing was tapered down in a stepwise fashion, until the patient arrived at the current dose of 7.5 mg BID Ruxolitinib, resulting in stable leukocyte counts around 25. As part of monitoring, we are currently conducting 6-month BM procedures and NGS for AML/MDS genes and Ruxolitinib resistance mutations alongside monthly CBCs. Our patient has been on Ruxolitinib for 9 months and has not developed any secondary contributing mutations and remains sensitive to therapy. We continue to monitor our patient’s response to Ruxolitinib to determine if germline predisposition alters the transformative nature of the disease. Within T618I germline CNL cases, we are investigating the difference in phenotype and disease burden that we observe in our patient’s family compared to literature. Based on current cases, it seems that germline CNL does not transform during childhood and adolescence into AML without the influence of a secondary mutation. Retrospective studies into previous cases of CNL to determine if there is a higher frequency of germline origin could pave the","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":"1 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41457654","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
T. Sarkar, Jennifer Lombardo, Shweta Singh, K. Gudmundsson, Holly Morris, Shyam Saran, J. Keller
{"title":"Abstract A55: Role of ID2 in acute myeloid leukemia progression and resistance to chemotherapy","authors":"T. Sarkar, Jennifer Lombardo, Shweta Singh, K. Gudmundsson, Holly Morris, Shyam Saran, J. Keller","doi":"10.1158/2643-3249.aml23-a55","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a55","url":null,"abstract":"\u0000 Acute myeloid leukemia (AML) is thought to be maintained by quiescent leukemia stem cells (LSCs), which are resistant to chemotherapy and responsible for relapse. Recent evidence suggests that a subset of LSCs with higher oxidative phosphorylation and/or increased senescence are responsible for resistance to chemotherapy and relapse. While several genes have been identified that are associated with the maintain LSC maintenance and promote resistance and relapse, the precious molecular mechanism(s) are currently not known. Inhibitor of DNA binding (ID) proteins are a group of Helix-Loop-Helix (HLH) transcription regulators, which function as dominant regulators of other HLH proteins (E proteins) that are required for normal hematopoietic, muscle, neuronal and other cell development. We found that loss of ID2 expression in murine hematopoietic cells results in activation, differentiation, and exhaustion of hematopoietic stem cells (HSCs). ID2 promotes quiescence by stabilizing HIF-1a expression. Analysis of TCGA data sets shows that high levels of ID2 expression is associated with reduced overall survival among AML patients, suggesting that ID2 may function to maintain LSCs. Similar to murine cells, knockdown (KD) of ID2 expression in human cord blood cells resulted in a loss of CD34+CD38− progenitors. Furthermore, knockdown of ID2 expression in the UT-7 AML cell line showed significantly reduced leukemogenic potential in NSG mice, suggesting that ID2 is required for AML cell growth in vivo. UT7-ID2-KD cell line shows significant reduction of colony number when cultured in low oxygen condition in vitro, suggesting a potential role of HIF protein. In comparison, overexpression (OE) of ID2 in the human AML cell line, MO7e, promotes their leukemic potential and burden in NSG mice in vivo, suggesting that ID2 may function to preserve LSC quiescence. Initial studies in primary AML-PDX cells showed reduced leukemic potential upon ID2 knock down. High level of ID2 expression is associated with poor chemotherapy response in patients. We found that MO7e-ID2-OE are resistance to Cytarabine (AraC), while UT7-ID2-KD are susceptible to AraC. Furthermore, we found that AML cell lines that survive AraC treatment show increased ID2 expression, suggesting that ID2 may promote survival of chemotherapy resistant AML cells. It has been reported that AraC resistance of AML cells is associated with increased senescence. We found that surviving UT7-ID2-KD cells after AraC treatment have significantly reduced senescence population compared to controls. Taken together, ID2 promotes normal murine and human HSC quiescence, and may function promote LSC maintenance and resistance to chemotherapy.\u0000 Citation Format: Tanmoy Sarkar, Jennifer Lombardo, Shweta Singh, Kristbjorn O Gudmundsson, Holly M Morris, Shyam Saran, Jonathan R Keller. Role of ID2 in acute myeloid leukemia progression and resistance to chemotherapy [abstract]. In: Proceedings of the AACR Special Conference: Ac","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":""},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48812548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kyle A. Romine, Daniel Ssozi, Tina Mujica, Y. Lee, Jennifer J. Trowbridge, P. van Galen
{"title":"Abstract A28: Mechanisms of adaptive immune evasion by pre-leukemic DNMT3A-mutated blood cell clones","authors":"Kyle A. Romine, Daniel Ssozi, Tina Mujica, Y. Lee, Jennifer J. Trowbridge, P. van Galen","doi":"10.1158/2643-3249.aml23-a28","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a28","url":null,"abstract":"\u0000 Hematopoietic stem cells (HSC) frequently acquire gene mutations with age. An expansion of blood cells derived from a single HSC is termed clonal hematopoiesis (CH), a condition that often precedes acute myeloid leukemia (AML). CH is common in elderly individuals: by age 60, approximately 10% of individuals have a clonally expanded blood cell population with a leukemia-associated mutation. The most common mutations in CH (50% of all cases) inactivate the DNA methyltransferase DNMT3A, which regulates gene expression via methylation of CpG rich regions. These mutations provide a selective advantage over healthy HSCs during aging and inflammation. Our goal is to discover mechanisms that allow for clonal expansion of DNMT3A-driven CH. A better understanding of these mechanisms will inspire new therapeutic strategies to limit clonal expansion and increase survival. To evaluate the transcriptional consequences of DNMT3A mutations in myeloid cells, we analyzed our previously published single-cell RNA-sequencing data of DNMT3A-mutated AML patient samples. We found a significant enrichment of antigen presentation signatures in DNMT3A mutated cells along the myeloid differentiation trajectory. CD8+ T cells from these same patients had significantly increased signatures of T cell activation and T cell exhaustion. T cell exhaustion occurs after an activated T cell has been continuously exposed to antigens. Exhausted T cells lack most effector functions and the ability to properly surveil (i.e. recognize mutant peptides presented on the surface of mutated cells and kill the target upon recognition) and restrain mutant cells from expanding. We hypothesize that DNMT3A-driven CH leads to escape of immune surveillance via chronic antigen stimulation and eventual promotion of T cell exhaustion. This would allow for further expansion of the mutated cells and the transformation of CH into AML. To functionally test this hypothesis, we evaluated whether a mouse model of Dnmt3a-CH recapitulated our human data. Indeed, scRNA-seq data of mouse Dnmt3a-CH cells showed significantly increased antigen presentation signatures across the myeloid differentiation trajectory. Additionally, CD8+ T cells had increased signatures of activation. We next functionally tested whether Dnmt3a-mutated CH cells had enhanced antigen presentation by pulsing bone marrow cells derived from Dnmt3a-CH mice with DQ-Ovalbumin, a self-quenched exogenous protein that becomes fluorescent when endocytosed and digested in the phagolysosome. Dnmt3a-mutated HSCs and LSKs had significantly increased DQ-OVA uptake and processing. In conclusion, we found that DNMT3A-CH cells have increased antigen presentation and processing which corresponds with enhanced immunogenicity. These findings support a model in which T cells constrain the expansion of DNMT3A-mutated HSCs, until T cell exhaustion occurs, leading to further expansion of the mutated clone and an increased chance of transformation into AML.\u0000 Citatio","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":""},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47751233","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yuki Nishida, Darah A Scruggs, Edward Ayoub, Lauren B Ostermann, Tallie Patsilevas, V. Ruvolo, Poo Yee Mak, B. Carter, S. Boettcher, A. Maiti, K. Sasaki, Qianxiang Zhou, Zhaohui Yang, H. Yan, Liandong Ma, M. Andreeff
{"title":"Abstract A31: C-MYC Targeting by Degradation: Novel Dual c-MYC/GSPT1 Degrader GT19715 Induces TP53-independent Cell Death in MYC-amplified Acute Myeloid Leukemia and Lymphomas","authors":"Yuki Nishida, Darah A Scruggs, Edward Ayoub, Lauren B Ostermann, Tallie Patsilevas, V. Ruvolo, Poo Yee Mak, B. Carter, S. Boettcher, A. Maiti, K. Sasaki, Qianxiang Zhou, Zhaohui Yang, H. Yan, Liandong Ma, M. Andreeff","doi":"10.1158/2643-3249.aml23-a31","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a31","url":null,"abstract":"\u0000 The oncoprotein c-MYC, a major regulator of the epigenome and transcriptome, is dysregulated in 70% of all human cancers. MYC is highly expressed in Burkitt lymphoma and TP53 mutant and venetoclax (ven) resistant AML (Sallman, Blood 2021, Nishida, ASH 2021). However, targeting c-Myc or the MYC pathway has not been successful and remains a major unmet clinical need. We developed the first cereblon E3 ligase modulators (CELMoDs) for c-MYC: GT19630 and GT19715 (salt form of GT19630). C-MYC was one of the top decreased proteins in chromatin-enriched proteomics, was pulled down by biotinylated GT19630 in in vitro affinity purification assay and was degraded with IC50 of 0.33 nM in MYC-amplified HL-60 cells. Proteasome inhibitor ixazomib completely blocked c-MYC reduction, suggesting a CRL4CRBN-dependent degradation. Blood cancer cell lines responded to GT19715 greater than other cancer cell lines such as lung, breast and brain tumors in a broad cell line panel, providing rationale to develop the degrader in hematologic malignancies. In agreement with other CELMoDs, proteomic analyses revealed degradation of translation termination factor GSPT1 (G1 to S phase transition proteins 1), an important factor in LSC survival Whereas a selective GSPT1 degrader CC-90009 reduced GSPT1 protein levels but not c-MYC, GT19715 reduced both c-MYC and GSPT1 and exerted a 20x higher cytoreduction than CC-90009 (IC50 of 1.8 nM vs 40.4 nM for GT19715 and CC-90009, respectively) in HL-60 cells. GT19630 degraded c-MYC and GSPT1 and inhibited tumor growth in a xenograft model with HL-60 cells. GT19715 eliminated circulating blasts and prolonged survival in the systemic Burkitt lymphoma model (Daudi). GT19715 significantly reduced human CD45+ AML blasts in peripheral blood, bone marrow (BM) and spleens compared to vehicle controls in vivo in a chemotherapy-resistant AML PDX model. MV4;11 venetoclax resistant (VR) cells demonstrated elevated protein levels of c-MYC and GSPT1 and GT19715 induced 4log10 cytoreduction in BM and prolonged survival of mice with MV4;11 VR cells. Baseline c-Myc protein levels associated with sensitivity to GT19715 in MOLM-13 cells with CRISPR engineered knockout/mutations of TP53 (R2 = 0.86, P = 0.02). GT19715 induced comparable cell death in primary AML samples with wild-type or mutant TP53 (95.4% and 91.7% cytoreduction, P = 0.48 for wild-type and mutant TP53 samples at 64 nM of GT19715, respectively). CD34+ AML cells were more susceptible to GT19715 than CD34- AML cells, suggesting a greater efficacy in AML stem/progenitor than in more mature AML cells. Notably, single cell mass cytometry revealed that CD34+ AML cells had higher c-MYC protein levels than CD34+ normal BM cells and GT19715 reduced c-Myc levels in CD34+ AML but not normal BM cells. GT19715 induced greater cytoreduction in CD34+ AML than in normal BM cells, suggesting a therapeutic window. Conclusions: The novel dual c-MYC/GSPT1 degrader GT19715 exerts TP53 independent preclinical a","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":""},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44570373","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pang-Ting Cheng, C. Teng, Y. Cheng, Mei-Chih Chen, Ho Lin
{"title":"Abstract A12: Androgen receptor inhibition is involved in Finasteride-reduced AML cell proliferation","authors":"Pang-Ting Cheng, C. Teng, Y. Cheng, Mei-Chih Chen, Ho Lin","doi":"10.1158/2643-3249.aml23-a12","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a12","url":null,"abstract":"\u0000 Acute myeloid leukemia (AML) is a top priority type of hematological malignancies to be studied and treated. Androgen receptor (AR) plays important roles in various cancers, such as types of the prostate, breast, liver, and lung. However, the role of AR in leukemia is so far uncertain. Finasteride is a 5 alpha-reductase inhibitor for reduction of cellular 5 alpha-dihydrotestosterone production and commonly used to treat benign prostatic hyperplasia for decades. Here, the data showed that Finasteride treatment significantly decreased the proliferation of AML cell lines, including KG-1 and HL-60. More specific to its impact on androgen’s actions, the authors found that Finasteride inhibited the phosphorylation and activation of AR, while the cell cycle-related proteins such as CDK1, Cyclin A, and p21 were unaffected. Interestingly, the increase of AKT phosphorylation was observed after Finasteride treatment, which is correspondent to the previous findings of mutual correlations between AR inhibition and AKT activation in prostate cancer cells. It implies that AR might play a physiological role in AML cells for proliferation, in which AR closely collaborates with AKT for a balance of regulation. In summary, these results suggest that Finasteride might reduce the proliferation of AML cell lines through AR inhibition. These findings could be helpful to uncover novel AML treatments in the future.\u0000 Citation Format: Pang-Ting Cheng, Chieh-Lin Jerry Teng, Yu-Chiao Cheng, Mei-Chih Chen, Ho Lin. Androgen receptor inhibition is involved in Finasteride-reduced AML cell proliferation [abstract]. In: Proceedings of the AACR Special Conference: Acute Myeloid Leukemia and Myelodysplastic Syndrome; 2023 Jan 23-25; Austin, TX. Philadelphia (PA): AACR; Blood Cancer Discov 2023;4(3_Suppl):Abstract nr A12.","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":""},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41996580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Benjamin Lim, C. Cher, J. Poh, K. Ngeow, Zi Yi Lim, Min-Han Tan
{"title":"Abstract A22: Complete mutation concordance with amplicon-based NGS between peripheral blood and bone marrow at 0.5% VAF threshold","authors":"Benjamin Lim, C. Cher, J. Poh, K. Ngeow, Zi Yi Lim, Min-Han Tan","doi":"10.1158/2643-3249.aml23-a22","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a22","url":null,"abstract":"\u0000 Background: Acute myeloid leukemia (AML) is a hematological malignancy characterized by genomic heterogeneity throughout disease progression and between patients. Traditionally, both multiparameter flow cytometry (MFC) and next-generation sequencing (NGS) samples are collected via bone marrow (BM) aspiration, which is an invasive procedure with risk of infection. Peripheral blood (PB) has been demonstrated as a good substitute for BM but the concordance at different levels of NGS variant allele frequency (VAF) has not been defined. This study aims to demonstrate the relationship of NGS signals between PB and BM.\u0000 Method: Genomic profiles of 60 matched samples of BM and PB were obtained retrospectively from 24 patients with de novo AML (n=17) or secondary AML (n=7). The samples were subjected to library preparation with a hematologic panel based on AmpliMark, an ultrasensitive amplicon-based NGS platform technology. The hematologic panel covers up to 45 genes common in myeloid and lymphoid neoplasms, with an established limit of detection of 0.1% VAF. Optimized bioinformatics pipeline and in-house sequencing noise removal algorithm were applied for variant selection. One of the secondary AML patients had a concurrent diagnosis of multiple myeloma; for the purposes of this analysis, the myeloma-associated variants were excluded.\u0000 Results: 50 out of 60 matched BM and PB samples (83.3%) in the cohort were positive for at least one variant in either sample type. Complete or partial concordance of genetic profiles was achieved in 86.7% of the matched BM and PB samples. All samples obtained during diagnosis or relapse showed complete concordance, while all partial concordant and discordant profiles were sampled during post-treatment or remission stages. All BM samples with ≥1.5% abnormal cells detected by MFC demonstrated complete concordance between BM and PB genetic profiles. Analysis at the individual variant level (n=115) showed 78.6% of positive percent agreement (PPA) comparing PB to BM in variant detection. For variants detected at ≥0.5% and ≥0.1% VAF in the BM samples, 100% PPA and 86.4% PPA were achieved from matched PB samples, respectively. A strong correlation of VAF (r=0.98) was also demonstrated between variants detected in BM and PB.\u0000 Conclusion: PB NGS is particularly sensitive for detection of myeloid specific mutations in de novo and secondary AMLs with complete concordance at VAFs ≥0.5%, when compared to BM. The high concordance and minimally invasive nature of PB sampling makes it a favorable option for diagnostic and monitoring of AML.\u0000 Citation Format: Benjamin Lim, Chae Yin Cher, Jonathan Poh, Kao Chin Ngeow, Zi Yi Lim, Min-Han Tan. Complete mutation concordance with amplicon-based NGS between peripheral blood and bone marrow at 0.5% VAF threshold [abstract]. In: Proceedings of the AACR Special Conference: Acute Myeloid Leukemia and Myelodysplastic Syndrome; 2023 Jan 23-25; Austin, TX. Philadelphia (PA): AACR; Blood Cancer Disco","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":""},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49654523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marlise L Guerrero Schimpf, Courtney C Sparger, Hsuan-Ting Huang, Dean Wade, Maria E. Figueroa
{"title":"Abstract A37: PDZD2 is essential for steady-state hematopoiesis and its 37-kDa secreted product, sPDZD2, functions as a soluble tumor suppressor in AML","authors":"Marlise L Guerrero Schimpf, Courtney C Sparger, Hsuan-Ting Huang, Dean Wade, Maria E. Figueroa","doi":"10.1158/2643-3249.aml23-a37","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a37","url":null,"abstract":"Abstract PDZ domain-containing protein 2 (PDZD2) is almost universally silenced through hypermethylation in myeloid malignancies. PDZD2 encodes for a 300kDa protein that it is cleaved into a 37kDa protein that is secreted to the tissue microenvironment (sPDZD2). Given that sPDZD2 has been reported to function as a tumor suppressor in solid tumors and its almost universal loss in AML, we hypothesized that PDZD2 is required for normal hematopoiesis and that sPDZD2 functions as a soluble tumor suppressor in the hematopoietic system. To determine if sPDZD2 is secreted to the bone marrow (BM) microenvironment we analyzed sPDZD2 levels in BM plasma isolated from primary AMLs and healthy donors using ELISA. We confirmed sPDZD2 secretion by normal CD34+ HSPC and detected lower levels in AMLs. In addition, treatment of a panel of AML cell lines (n=9) and primary human AML cells (n=4) with recombinant sPDZD2 (r-sPDZD2; dose: 100nM-300nM/day) led to growth inhibition, upregulation of differentiation markers (CD15/CD11b) and cell cycle arrest in G0/G1. To identify signaling pathways downstream of sPDZD2, we treated MV4-11 cells with 100nM r-sPDZD2 and performed phospho-kinase protein array followed by immunoblot. In vitro treatment with r-sPDZD2 led to inhibition of CREB(S133) phosphorylation, as well as increased GSK3b(S9), RSK1/2(S221/S227) and p53(S46) phosphorylation. In order to determine whether PDZD2 plays a role in normal hematopoiesis we performed in vitro differentiation in CRISPR-edited human HSPCs. We used a Cy3-labeled recombinant Cas9 protein and specific single guide RNAs (gRNAs) targeted against PDZD2. Cy3-positive cells were then used for in vitro myeloid or erythroid differentiation, and cell surface markers were evaluated by flow-cytometry analysis over a period of 11 days. While PDZD2-edited cells (PDZD2 KO) displayed a minor defect on myeloid differentiation due to insufficient upregulation of CD15, erythropoiesis appears to be more compromised, with a significant failure to adequately upregulate both CD235a and CD7. To determine the role of Pdzd2 in vivo, we took advantage of a Pdzd2Gt/Gt gene-trap knock-out mouse model and performed peripheral blood (PB) and BM analysis. PB counts at 8-weeks of age showed anemia, with a significant decrease in both red blood cell counts and hemoglobin levels, an observation compatible with the impaired erythropoiesis seen in vitro after PDZD2 KO in human HSPCs. In addition, we observed a trend to decreased MEPs and short-term HSCs, in the BM of 12-week-old mice (n=3), though the small size of the cohort prevented robust statistical analysis. In summary, our findings are the first to shed light on a previously unrecognized role for PDZD2 in hematopoiesis. Loss of PDZD2 in HSPC resulted in impaired erythroid differentiation both in vitro and in vivo. Moreover, our findings validate a soluble tumor suppressor role for sPDZD2 in AML, similar to that seen in solid tumors. Importantly, we further demonstrate","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":"15 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"136095170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xufeng Chen, Qiao Lu, Hua Zhou, Jia Liu, B. Nadorp, Audrey Lasry, Zhengxi Sun, Jiangyan Zhang, M. Cammer, Kun Wang, Zoe B. Ciantra, J. You, Qianjin Guo, Hongbing Zhang, Debrup Sengupta, Ahmad Boukhris, Cheng Liu, P. Cresswell, P. Dahia, Jun Wang, I. Aifantis
{"title":"Abstract A10: Targeting MHC-I antigen presentation for cancer immune evasion in acute myeloid leukemia","authors":"Xufeng Chen, Qiao Lu, Hua Zhou, Jia Liu, B. Nadorp, Audrey Lasry, Zhengxi Sun, Jiangyan Zhang, M. Cammer, Kun Wang, Zoe B. Ciantra, J. You, Qianjin Guo, Hongbing Zhang, Debrup Sengupta, Ahmad Boukhris, Cheng Liu, P. Cresswell, P. Dahia, Jun Wang, I. Aifantis","doi":"10.1158/2643-3249.aml23-a10","DOIUrl":"https://doi.org/10.1158/2643-3249.aml23-a10","url":null,"abstract":"\u0000 Acute Myeloid Leukemia (AML) is a clonal hematopoietic neoplasm, accounting for ~80% of acute leukemia cases in adults. The poor clinical outcome of AML patients (~27% five-year overall survival) is largely due to the inefficiency and toxicity of standard therapies. In the past decade, immunotherapies or cellular therapies (CAR-T/TCR-T cells) have achieved FDA approvals in certain types of solid tumors and leukemia, but currently there is no approved immunotherapy in AML. One potential reason is that AML actively inhibits antigen presentation (AP) to reduce its immunogenicity, thus dampening the efficacies of immunotherapies. This growing consensus is supported by the clinical evidence that AML exhibits low levels of AP machinery at diagnosis and even lower when relapsed from conventional chemotherapies. Besides, better outcomes could be achieved when combining immune checkpoint blockade with neo-antigens-inducing hypomethylating agents, highlighting the importance of AP in AML. We thus hypothesize that targeting tumor-endogenous AP suppressors will enhance AML immunogenicity and benefit immunotherapies.\u0000 Recent studies have suggested that autophagy or soluble protein PCSK9 can mediate MHC-I degradation or disrupt MHC-I recycling, respectively. Transcription/epigenetic repressors TRAF3 and EZH2, as well as thymidylate synthase, were identified as MHC-I inhibitors. However, their specificity in MHC-I modulation and precise roles in regulating tumor antigenic peptide-MHC-I complexes (pMHC-I) are still unclear. Hence, there is an increasing need to identify AML-specific AP regulation mechanisms.\u0000 To map such mechanisms, we performed pMHC-I-guided CRISPR screens in both human and mouse AML cell lines. Through these screens, we constructed positive and negative regulatory networks of pMHC-I modulation and compared the role of these novel regulators in the simultaneous modulation of MHC-I expression. This is the first-in-class systematic identification of AP regulators in AML. Among these negative AP regulators, we are particularly interested in the interferon regulatory factor 2 binding protein 2 (IRF2BP2), an AML-specific transcriptional regulator that is highly expressed in AML compared with normal hematopoietic stem and progenitor cells (HSPCs). Notably, IRF2BP2 expression negatively correlates with major histocompatibility complex class I (MHC-I) expression, interferon (IFN) response signatures, and T cell activity in AML patients. Ablation of IRF2BP2 enhanced AP in AML and facilitated T cell-mediated elimination of AML. Moreover, IRF2BP2 depletion synergized with IFN treatment to further boost AP at both transcriptional and protein levels. Our findings reveal a new class of tumor-associated immune-evasion mechanisms that target AP, with potential application as therapeutic targets for next-generation cancer immunotherapies.\u0000 Citation Format: Xufeng Chen, Qiao Lu, Hua Zhou, Jia Liu, Bettina Nadorp, Audrey Lasry, Zhengxi Sun, Jiangyan Zhang, Mich","PeriodicalId":29944,"journal":{"name":"Blood Cancer Discovery","volume":" ","pages":""},"PeriodicalIF":11.2,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45233178","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}