scholarly journals Antibody-based therapy of leukaemia

Author(s):  
John C. Morris ◽  
Thomas A. Waldmann

Over the past decade, monoclonal antibodies have dramatically impacted the treatment of haematological malignancies, as evidenced by the effect of rituximab on the response rate and survival of patients with follicular and diffuse large B cell non-Hodgkin's lymphoma. Currently, only two monoclonal antibodies – the anti-CD33 immunotoxin gemtuzumab ozogamicin and the CD52-directed antibody alemtuzumab – are approved for treatment of relapsed acute myeloid leukaemia in older patients and B cell chronic lymphocytic leukaemia, respectively. Although not approved for such treatment, alemtuzumab is also active against T cell prolymphocytic leukaemia, cutaneous T cell lymphoma and Sézary syndrome, and adult T cell leukaemia and lymphoma. In addition, rituximab has demonstrated activity against B cell chronic lymphocytic and hairy cell leukaemia. Monoclonal antibodies targeting CD4, CD19, CD20, CD22, CD23, CD25, CD45, CD66 and CD122 are now being studied in the clinic for the treatment of leukaemia. Here, we discuss how these new antibodies have been engineered to reduce immunogenicity and improve antibody targeting and binding. Improved interactions with Fc receptors on immune effector cells can enhance destruction of target cells through antibody-dependent cellular cytotoxicity and complement-mediated cell lysis. The antibodies can also be armed with cellular toxins or radionuclides to enhance the destruction of leukaemia cells.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2437-2437 ◽  
Author(s):  
Alla Dolnikov ◽  
Shen Sylvie ◽  
Ning Xu ◽  
Tracey O'Brien

Abstract Allogeneic stem cell transplantation is a curative therapy for refractory, relapsed and high risk leukaemia and lymphoma. Despite improvements in leukaemia treatment the disease relapse occurs in 30-50% of patients and remains unacceptably high. There is thus a clear need for novel agents in the treatment of leukaemia relapse. Leukaemia specific T cells genetically modified with Chimeric Antigen Receptor (CAR) generated in the laboratory to target malignant cells is a novel approach with proven success in early phase human trials. Patient's own T cells can be genetically modified in the laboratory to target tumour associated antigens through the introduction of CAR. CAR-modified T cells are amplified ex vivo to numbers suitable for adoptive cell therapy and administered to the patient upon preconditioning. Unfortunately, CAR-modified mature T cells often rapidly differentiate into short-lived effector cells that exhibit limited anti-tumour activity in vivo. Here we propose to use CAR-modified haematopoietic stem cells (HSC) to generate CART- cells. We hypothesise that CAR-modified HSC will provide long-lasting supply of CAR-T cells mediating sustained anti-tumour activity. Umbilical cord blood-derived CD34+ cells enriched with HSC were genetically modified to express CAR targeting CD19 antigen(CAR19) widely expressed on B-cell malignancies. An optimised gamma-retroviral gene transfer method was used to transduce CD34+ stem cells with CAR19. CAR19-transduced stem cells generate CD33+ myeloid, CD56+ natural killer and CD34-CD7+ T cell precursors expressing CAR19 and undergo normal proliferation and differentiation in vitro in colony forming unit (CFU) assay. B cell development analysed in co-cultures with bone marrow stroma MS5 cells, however, was completely suppressed. CAR19-transduced CD34+ stem cells were transplanted to immunocompromised NOD-SCID IL2Rg-/- (NSG) mice to generate multilineage immune effector cells exhibited delayed early engraftment. It is relevant that CD19+ B cells regenerate first in this mouse model. Thus mice transplanted with CAR19-transduced stem cells exhibited severe CD19+ B cell depletion confirming the functionality of CAR19 expressed in human B cells. To promote T cell regeneration, CD34+ HSCs were co-cultured with Notch ligand DL4 for 1 week before infusion. CD3+T cells were detectable in the blood of mice infused with DL4-pre-cultured CD34+ HSCs as soon as 6 weeks post-transplant while control CD34+ stem cells that were not co-cultured without DL4 exhibited significantly delayed T cell regeneration. Importantly, reduced leukaemia burden was observed in mice reconstituted with CAR19-modified stem cells and infused with human CD19+ leukaemia NALM6 cells. These data proves the feasibility of the stem cell approach to generate potent immune effector cells capable of fighting cancer. Further analysis aimed to identify the role of different multilineage immune effector cells in mediating anti-tumour activity in stem cell reconstituted mice is currently being conducted. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 1 (1) ◽  
pp. 7-12 ◽  
Author(s):  
David Zahavi ◽  
Dalal AlDeghaither ◽  
Allison O’Connell ◽  
Louis M Weiner

ABSTRACT The targeting of surface antigens expressed on tumor cells by monoclonal antibodies (mAbs) has revolutionized cancer therapeutics. One mechanism of action of antibody-based immunotherapy is the activation of immune effector cells to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). This review will summarize the process of ADCC, its important role in the efficacy of mAb therapy, how to measure it, and finally future strategies for antibody design that can take advantage of it to improve clinical performance.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2538-2538
Author(s):  
Joerg M. Aswald ◽  
Xing-Hua Wang ◽  
Sandra Aswald ◽  
Loralyn A. Benoit ◽  
Mark Minden ◽  
...  

Abstract Prolonging event-free survival of AML with autologous activated immune cells is a promising concept. GD-T cells are a rare circulating lymphocyte population (1%) and a component of the innate immune system capable of exerting anti-neoplastic activity. Their role as potential anti-cancer immune effector cells deserves further exploration. It is noteworthy that GD-T cells are over-represented in reactive regions surrounding melanoma lesions. While patients with an accumulation of GD-T cells showed a survival benefit over those who did not, such increases were not present in patients with metastatic disease and high tumor cell burden (Bachelez, J. Invest. Dermatol.98:369,1992). Little is known about the role of GD-T cells as immuno-effectors, their absolute numbers in peripheral blood or the feasibility of purifying functional GD-T cells from patients with AML. We are interested in testing the clinical feasibility of using GD-T cells freshly purified from PB against minimal residual disease in AML. As a first step towards achieving this goal, we compared circulating GD-T cell levels sequentially in 33 AML patients with 20 healthy adult volunteers. We used ultra-low volume multi-color flow-cytometry and microbeads to measure absolute numbers of GD-T cells in PB. Functional studies were done by the chromium release assay and single-cell intra-cellular interferon-gamma detection. We observed that AML patients with a high leukemic blast cell burden (e.g. prior to chemotherapy) had marginally decreased GD-T cell levels compared with healthy controls: median 38/μl, Q1-Q3, 27–86/μl, versus median 83/μl, Q1-Q3, 45–122/μl, respectively, p= 0.051. We re-examined the AML patients at several time points after induction therapy and observed significantly increased numbers of GD-T cells in patients with lower but detectable residual disease (either molecular maker positive or borderline bone marrow blast infiltration by morphology) compared to patients with persistently high blast cell burden: median 105/μl, Q1-Q3, 105–133/μl versus median, 7/μl, Q1-Q3, 6–15/μl; p=0.008. Patients with residual disease also showed significantly higher numbers of absolute GD-T cells per microliter blood compared to those retested after they had achieved complete remission (CR); p=0.0025. In CR, GD-T cell counts remained lower than those of healthy individuals: median 33/μl, Q1-Q3, 22–35/μl versus median 83/μl, Q1-Q3, 45–122/μl; p=0.030. Interestingly, we found a sharp increase (on average, 4.9-fold higher than values obtained in CR) in GD-T levels at the time of very early morphologic (n=3) or molecular relapse (n=2). Hence, we were interested in studying the functional properties of the GD-T cells from AML patients. We were able to isolate functional GD-T cells from the PB of patients with AML in CR-1 in sufficient numbers and purity to assay for interferon-gamma and found that similar numbers of GD-T cells expressed the Th1 cytokine compared with healthy controls: 84% versus 93% of all GD-T cells, respectively. We also showed that GD-T cells were able to kill leukemic target cells (AML-OCI2) in vitro more efficiently than CD3+ T cells. Our data suggest that further studies to investigate the potential therapeutic role of autologous GD-T cells in patients with AML in CR are warranted.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e22198-e22198
Author(s):  
Mohammad Hojat Farsangi ◽  
Fatemeh Ghaemimanesh ◽  
Amir Hossein Daneshmanesh ◽  
Ali-Ahmad Bayat ◽  
Jafar Mahmoudian ◽  
...  

e22198 Background: The receptor tyrosine kinase ROR1 has important functions during normal embryogenesis. It is overexpressed and of importance for the survival of different malignancies, including lung adenocarcinoma, breast cancer and chronic lymphocytic leukemia (CLL). ROR1 inhibition in CLL cells and cell lines with high expression of ROR1 induced specific apoptosis of the cells. There is however little information on the expression and function of ROR1 in melanoma. Here, we investigated ROR1 expression, phosphorylation and the effects of anti-ROR1 monoclonal antibodies (mAbs) and ROR1 suppressing siRNA on seven melanoma cell lines. Methods: ROR1 expression and phosphorylation was analysed in a series of malignant melanoma cell lines using RT-PCR, immunocytochemistry, flow cytometry and western blot. Annexin V/PI staining, MTT assay, PARP and caspase 8 and 9 cleavage as well as MCL-1 protein were used for detection of apoptosis induced by anti-ROR1 mAbs. The cytotoxic effects of anti-ROR1 mAbs were evaluated in the absence or presence of complement (CDC) or immune effector cells (ADCC). Transfection of melanoma cell lines using ROR1 siRNA was used for gene silencing. Results: ROR1 was shown to be overexpressed to a varying degree at protein level. ROR1 was phosphorylated at tyrosine and serine residues. Three out of four anti-ROR1 mAbs (clones 3H9, 5F1 and 1A8) induced a significant direct apoptosis of the ESTDAB049, ESTDAB112, DFW and A375 cell lines as well as cell death in the presence of complement or immune effector cells. ESTDAB081 and 094 cell lines were resistant to direct apoptosis of the four anti-ROR1 mAbs alone compared to the other melanoma cell lines, but not to complement dependent cytotoxicity (CDC) and antibody dependent cell mediated cytotoxicity (ADCC). ROR1 siRNA transfection induced down-regulation of the ROR1 expression both at the mRNA and protein levels proceeded by a significant apoptosis of the melanoma cells. Conclusions: The results indicate that ROR1 may play an important role in the survival of melanoma cells. The surface expression of ROR1 on melanoma cells may support the notion that ROR1 might be a suitable targeted for mAb therapy.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3969-3969
Author(s):  
Xiaoyan Feng ◽  
Yue Song ◽  
Zhaoming Li ◽  
Xudong Zhang ◽  
Lugui Qiu ◽  
...  

Introduction Extranodal NK/T cell lymphoma (NKTCL) is an aggressive malignance that is correlated closely with persistent Epstein-Barr virus (EBV) infection and belongs to a latency II EBV disease. Chronic persistent viral infection is known to result in T cell exhaustion, which will dramatically impede anti-tumor immunity. Delineating mechanisms of immunosuppression contributes to the development of novel immuno-therapeutically strategies. Herein, we described the immune status and possible role of latent membrane protein 1(LMP1) in NKTCL. Methods Peripheral blood mononuclear cells (PBMCs) from newly diagnosed NKTCL patients and age-matched healthy donors (HDs) were isolated and stained with surface marker or intracellular marker after permeabilization. The frequency of EBV-specific cytotoxic T cells (CTLs) was analyzed by staining with HLA-A2 tetramers assembled with synthetic peptides from LMP1. T cell proliferation was detected by dilution of stained CFSE, and apoptosis of T cells was performed via annexinV and 7-AAD dual-staining. Cytotoxicity assay of natural killer cells (NK) was measured by detecting apoptosis of CFSE-labeled K562 cells after co-culturing with PBMCs. All flow cytometry was performed by BD FACS CantoII, and analyzed with FlowJo software. Results PBMCs from 19 NKTCL patients and 18 HDs were analyzed, which showed that the patients had higher ratio of CD4+/CD8+ cells and lower frequency of CD3-CD56+NK cells, higher percentage of immunosuppressive CD4+CD25hiCD127low T regulatory cells (Tregs) and HLA-DR-CD11b+CD33+ myeloid derived suppressor cells (MDSCs). Notably, the ratio of CD8+/Tregs, a parameter representing immune effector cells status, was obviously decreased in NKTCL patients (Figure-a). Given the potential association of T cell exhaustion and NKTCL, we detected expression of T cell exhaustion markers on T cells from NKTCL patients, and found that both CD4+ and CD8+ T cells expressed much higher level of inhibitory molecules PD1, CTLA4, TIGIT and TIM3 in NKTCL patients than that of HDs (Figure-b). To further explore cellular immunity in NKTCL, T cells were divided into four subsets: CD45RA-CCR7+ central memory T cells (Tcm), CD45RA+CCR7+ terminally differentiated effector T cells (Temra), CD45RA+CCR7- naïve T cells (Tn), CD45RA-CCR7- effector memory T cells (Tem) (Figure-c). It showed that CD4+ T cells had lower Tcm, Temra and Tn but much higher proportion of Tem compared with HDs (Figure-d). Moreover, we found that expression of PD1 and CTLA4 were upregulated on all lymphocyte subsets in NKTCL patients than HDs (Figure-e). We collected additional 13 NKTCL patients and 10 HDs with positive HLA-A2 phenotype to measure percentages of EBV-specific CTLs, which showed that frequency of EBV-specific CTLs was remarkably decreased in NKTCL patients (Figure-f). In addition, EBV-specific CTLs from NKTCL patients were more likely to express higher levels of exhaustion markers but produced much less IFN-γ (Figure g-h). To explore the potent mechanism of immunosuppression in NKTCL, LMP1 attracted our attention, which had been proven capable of activating multiple signaling pathways to exert its oncogenesis function. To elucidate effect of LMP1 on immune cells, we constructed two LMP1-derived peptides and found that LMP1-derived peptides suppressed the proliferation of CD4+ and CD8+ T cells and inhibited IFN-γ production of CD8+ T cells and NK cells (Figure i-j). Meanwhile, LMP1 promoted apoptosis of both CD4+ and CD8+ T cells (Figure-k). Subsequently regulation of LMP1 on exhaustion markers of T cells were detected, which showed that PD1, CTLA4, TIGIT and TIM3 were significantly upregulated on both CD4+ and CD8+ T cells after treatment with LMP1 derived peptides (Figure-m). Furthermore, LMP1 impaired NK cytotoxicity ability, evidenced by decrease apoptosis of target cells (Figure-n). Besides its effect on immune effector cells, we also found that LMP1-expressing NKTCL cell lines obviously induced both Tregs and MDSCs after co-culture. Interestingly, the extent of induction by LMP1 of Tregs and MDSCs were in line with the expression level of LMP1 on NKTCL cells (Figure o-p). Conclusion Our study showed that LMP1 contributes to deficient cellular immunity in NKTCL patients, providing us with more insight into immunosuppressive in this entity of disease, which would lead to identification of novel treatment strategies. Figure Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 288 (23) ◽  
pp. 16334-16347 ◽  
Author(s):  
Franziska Stehle ◽  
Kristin Schulz ◽  
Corinna Fahldieck ◽  
Jana Kalich ◽  
Rudolf Lichtenfels ◽  
...  

The multikinase inhibitors sunitinib, sorafenib, and axitinib have an impact not only on tumor growth and angiogenesis, but also on the activity and function of immune effector cells. In this study, a comparative analysis of the growth inhibitory properties and apoptosis induction potentials of tyrosine kinase inhibitors on T cells was performed. Tyrosine kinase inhibitor treatment resulted in a dramatic decrease in T cell proliferation along with distinct impacts on the cell cycle progression. This was at least partially associated with an enhanced induction of apoptosis although triggered by distinct apoptotic mechanisms. In contrast to sunitinib and sorafenib, axitinib did not affect the mitochondrial membrane potential (Δψm) but resulted in an induction or stabilization of the induced myeloid leukemia cell differentiation protein (Mcl-1), leading to an irreversible arrest in the G2/M cell cycle phase and delayed apoptosis. Furthermore, the sorafenib-mediated suppression of immune effector cells, in particular the reduction of the CD8+ T cell subset along with the down-regulation of key immune cell markers such as chemokine CC motif receptor 7 (CCR7), CD26, CD69, CD25, and CXCR3, was not observed in axitinib-treated immune effector cells. Therefore, axitinib rather than sorafenib seems to be suitable for implementation in complex treatment regimens of cancer patients including immunotherapy.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e13559-e13559
Author(s):  
Tsuyoshi Mori ◽  
Satoshi Wada ◽  
Kelly Olino ◽  
Barish H Edil ◽  
Drew M. Pardoll ◽  
...  

e13559 Background: Chemotherapy has been shown to act as an immune response modifier. Many groups are using chemotherapy to induce immune responses such as by using low doses of cyclophosphamide. Building up on our previous work (Murata et al., 2006, Stearns et al., 2011), we demonstrate that intraductal (i.duc), but not the intravenous (IV) route of injection of chemotherapy to HER2/neu transgenic mouse mammary glands (MMG) alters the tumor environment to effectively induce immune effector cells. Methods: 1.Tumor-free survival: 5-fluorouracil (5FU) was administered intraductally to the MMGs on the left side of mice; the MMG on right side were untreated. 2. FACS analysis: 20w mice were administered 5FU 2 times in a 4 week interval i.duc only to the left side, IV, or no treatment (NT). At week 5, the mice were sacrificed and the regional lymph nodes (RLN) and spleen were removed. Lymphocytes from RLN and spleen were analyzed. 3. Tumor re-challenge: GT8 cancer cells were injected into the one side of 4th mammary gland fat pad of 12 week mice. The mice were then injected i.duc with 5-FU to all teats in the tumor implanted side, or IV or given NT. The surviving mice were re-challenged with GT8 cancer cells into the ipsilateral 3rd mammary gland fat pad, 4 weeks after the first inoculation. Results: 1. The tumor incidence in the 5FU treated side after 16 weeks was significantly lower (13% of 70 glands) compared to mice that received NT (34% of 150, p=0.001), and IV (28% of 70, p=0.03). The tumor incidence in the untreated side of 5FU-treated mice was also significantly lower (18% of 70) compared to the NT (p=0.01). 2. The number of CD8 T cell showed no change among the groups in the RLN but was significantly lower (p=0.01) in the spleen of the i.duc. The number of CD62LLOW+ T cell in treated side of RLN was significantly higher compared to the IV (p=0.02) and NT (p=0.02). The number of CD62LLOW+ was low in the spleen. 3. The survivors in the group previously treated with i.duc 5-FU were able to achieve a lasting rejection of the tumor re-challenge when compared to mice who previously received 5-FU IV. Conclusions: I.duc administration of 5FU into MMG effectively induced immune effector cells and prevented mammary tumor growth in HER2/neu transgenic mice.


2021 ◽  
Vol 9 (2) ◽  
pp. e001691
Author(s):  
Kellsye P Fabian ◽  
Michelle R Padget ◽  
Rika Fujii ◽  
Jeffrey Schlom ◽  
James W Hodge

BackgroundDifferent types of tumors have varying susceptibility to immunotherapy and hence require different treatment strategies; these cover a spectrum ranging from ‘hot’ tumors or those with high mutational burden and immune infiltrates that are more amenable to targeting to ‘cold’ tumors that are more difficult to treat due to the fewer targetable mutations and checkpoint markers. We hypothesized that an effective anti-tumor response requires multiple agents that would (1) engage the immune response and generate tumor-specific effector cells; (2) expand the number and breadth of the immune effector cells; (3) enable the anti-tumor activity of these immune cells in the tumor microenvironment; and (4) evolve the tumor response to widen immune effector repertoire.MethodsA hexatherapy combination was designed and administered to MC38-CEA (warm) and 4T1 (cool) murine tumor models. The hexatherapy regimen was composed of adenovirus-based vaccine and IL-15 (interleukin-15) superagonist (N-803) to engage the immune response; anti-OX40 and anti-4-1BB to expand effector cells; anti-PD-L1 (anti-programmed death-ligand 1) to enable anti-tumor activity; and docetaxel to promote antigen spread. Primary and metastatic tumor growth inhibition were measured. The generation of anti-tumor immune effector cells was analyzed using flow cytometry, ELISpot (enzyme-linked immunospot), and RNA analysis.ResultsThe MC38-CEA and 4T1 tumor models have differential sensitivities to the combination treatments. In the ‘warm’ MC38-CEA, combinations with two to five agents resulted in moderate therapeutic benefit while the hexatherapy regimen outperformed all these combinations. On the other hand, the hexatherapy regimen was required in order to decrease the primary and metastatic tumor burden in the ‘cool’ 4T1 model. In both models, the hexatherapy regimen promoted CD4+ and CD8+ T cell proliferation and activity. Furthermore, the hexatherapy regimen induced vaccine-specific T cells and stimulated antigen cascade. The hexatherapy regimen also limited the immunosuppressive T cell and myeloid derived suppressor cell populations, and also decreased the expression of exhaustion markers in T cells in the 4T1 model.ConclusionThe hexatherapy regimen is a strategic combination of immuno-oncology agents that can engage, expand, enable, and evolve the immune response and can provide therapeutic benefits in both MC38-CEA (warm) and 4T1 (cool) tumor models.


2021 ◽  
Vol 79 (3) ◽  
pp. 961-968
Author(s):  
Wolfgang J. Streit ◽  
Habibeh Khoshbouei ◽  
Ingo Bechmann

Microglia constitute the brain’s immune system and their involvement in Alzheimer’s disease has been discussed. Commonly, and in line with the amyloid/neuroinflammation cascade hypothesis, microglia have been portrayed as potentially dangerous immune effector cells thought to be overactivated by amyloid and producing neurotoxic inflammatory mediators that lead to neurofibrillary degeneration. We disagree with this theory and offer as an alternative the microglial dysfunction theory stating that microglia become impaired in their normally neuroprotective roles because of aging, i.e., they become senescent and aging neurons degenerate because they lack the needed microglial support for their survival. Thus, while the amyloid cascade theory relies primarily on genetic data, the dysfunction theory incorporates aging as a critical etiological factor. Aging is the greatest risk factor for the sporadic (late-onset) and most common form of Alzheimer’s disease, where fully penetrant genetic mutations are absent. In this review, we lay out and discuss the human evidence that supports senescent microglial dysfunction and conflicts with the amyloid/neuroinflammation idea.


Sign in / Sign up

Export Citation Format

Share Document