1
|
Fan S, Wang T, You F, Zhang T, Li Y, Ji C, Han Z, Sheng B, Zhai X, An G, Meng H, Yang L. B7-H3 chimeric antigen receptor-modified T cell shows potential for targeted treatment of acute myeloid leukaemia. Eur J Med Res 2023; 28:129. [PMID: 36941687 PMCID: PMC10026503 DOI: 10.1186/s40001-023-01049-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/07/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND AND AIMS Chimeric antigen receptor (CAR)-T cell therapy is a novel type of immunotherapy. However, the use of CAR-T cells to treat acute myeloid leukaemia (AML) has limitations. B7-H3 is expressed in several malignancies, including some types of AML cells. However, its expression in normal tissues is low. Therefore, B7-H3 is ideal for targeted AML therapy. MATERIALS AND METHODS First, we constructed B7-H3 CAR that can target B7-H3, and then constructed B7-H3-CAR-T cells in vitro, which were co-incubated with six AML cell lines expressing different levels of B7-H3, respectively. The toxicity and cytokines were detected by flow cytometry. In vivo, AML model was established in B-NSG mice to study the toxicity of B7-H3-CAR T on AML cells. RESULTS In vitro functional tests showed that B7-H3-CAR-T cells were cytotoxic to B7-H3-positive AML tumor cells and had good scavenging effect on B7-H3-expressing AML cell lines, and the cytokine results were consistent. In vivo, B7-H3-CAR-T cells significantly inhibited tumor cell growth in a mouse model of AML, prolonging mouse survival compared with controls. CONCLUSION B7-H3-CAR-T cells may serve as a novel therapeutic method for the targeted treatment of AML.
Collapse
Affiliation(s)
- Shuangshuang Fan
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Tian Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
| | - Fengtao You
- PersonGen BioTherapeutics (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
| | - Tingting Zhang
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yafen Li
- PersonGen BioTherapeutics (Suzhou) Co., Ltd., Suzhou, Jiangsu, China
| | - Cheng Ji
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Zhichao Han
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Binjie Sheng
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xiaochen Zhai
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Gangli An
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Huimin Meng
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China.
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.
| | - Lin Yang
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, 215123, Jiangsu, China.
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China.
- PersonGen BioTherapeutics (Suzhou) Co., Ltd., Suzhou, Jiangsu, China.
| |
Collapse
|
2
|
Sioud M, Pettersen S, Ailte I, Fløisand Y. Targeted Killing of Monocytes/Macrophages and Myeloid Leukemia Cells with Pro-Apoptotic Peptides. Cancers (Basel) 2019; 11:cancers11081088. [PMID: 31370273 PMCID: PMC6721331 DOI: 10.3390/cancers11081088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 01/12/2023] Open
Abstract
Several cells of myeloid origin, such as monocytes and macrophages are involved in various human disorders, including cancer and inflammatory diseases. Hence, they represent attractive therapeutic targets. Here we developed three lytic hybrid peptides, by fusing a monocyte- and macrophage-binding peptide to pro-apoptotic peptides, and investigated their killing potency on blood monocytes, macrophages, and leukemia cells. We first showed that the targeting NW peptide is effective for depleting monocytes from whole peripheral blood mononuclear cells (PBMCs). Incubating the cells with biotin-conjugated NW peptide, and the subsequent capture on streptavidin-conjugated magnetic beads, depleted monocytes from the PBMCs. The NW peptide also depleted myeloid leukemia blasts from patient PBMCs. The treatment of the PBMCs with the lytic hybrid NW-KLA peptide killed monocytes, but not lymphocytes and primary mammary epithelial cells. Additionally, the fusion peptide exhibited a potent toxicity against macrophages and leukemia cells. The free lytic KLA peptide did not affect cells. Similarly, a second lytic hybrid peptide killed macrophages, leukemia cell lines, and blood leukemia blasts from patients with acute and chronic myeloid leukemia. The IC50 towards target cells were in the low macromolar range (4–12 µM). Overall, the data indicate that the NW peptide could be a potential drug delivery agent for monocytes, macrophages, and leukemia cells. Moreover, the engineered lytic hybrid peptides acting alone, or in combination with other therapeutic agents, might benefit many cancer patients and overcome drug resistance.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, N0379 Oslo, Norway.
| | - Solveig Pettersen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, N0379 Oslo, Norway
| | - Ieva Ailte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, N0379 Oslo, Norway
| | - Yngvar Fløisand
- Department of Haematology, Oslo University Hospital-Rikshospitalet, Sognsvannvien 20, N0372 Oslo, Norway
| |
Collapse
|
3
|
Guinn BA, Mohamedali A, Mills KI, Czepulkowski B, Schmitt M, Greiner J. Leukemia Associated Antigens: Their Dual Role as Biomarkers and Immunotherapeutic Targets for Acute Myeloid Leukemia. Biomark Insights 2017. [DOI: 10.1177/117727190700200015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Leukemia associated antigens (LAAs) are being increasingly identified by methods such as cytotoxic T-lymphocyte (CTL) cloning, serological analysis of recombinant cDNA expression libraries (SEREX) and mass spectrometry (MS). In additional, large scale screening techniques such as microarray, single nucleotide polymorphisms (SNPs), serial analysis of gene expression (SAGE) and 2-dimensional gel electrophoresis (2-DE) have expanded our understanding of the role that tumor antigens play in the biological processes which are perturbed in acute myeloid leukemia (AML). It has become increasingly apparent that these antigens play a dual role, not only as targets for immunotherapy, but also as biomarkers of disease state, stage, response to treatment and survival. We need biomarkers to enable the identification of the patients who are most likely to benefit from specific treatments (conventional and/or novel) and to help clinicians and scientists improve clinical end points and treatment design. Here we describe the LAAs identified in AML, to date, which have already been shown to play a dual role as biomarkers of AML disease.
Collapse
Affiliation(s)
- Barbara-ann Guinn
- Department of Haematological Medicine, King's College London School of Medicine, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU
| | - Azim Mohamedali
- Department of Haematological Medicine, King's College London School of Medicine, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU
| | - Ken I. Mills
- Department of Haematology, University Hospital of Wales, Heath Park, Cardiff, CF4 4XN, U.K
| | - Barbara Czepulkowski
- Department of Haematological Medicine, King's College London School of Medicine, The Rayne Institute, 123 Coldharbour Lane, London, SE5 9NU
| | - Michael Schmitt
- Third Clinic for Internal Medicine, University of Ulm, Germany
| | - Jochen Greiner
- Third Clinic for Internal Medicine, University of Ulm, Germany
| |
Collapse
|
4
|
Hofmann S, Mead A, Malinovskis A, Hardwick NR, Guinn BA. Analogue peptides for the immunotherapy of human acute myeloid leukemia. Cancer Immunol Immunother 2015; 64:1357-67. [PMID: 26438084 PMCID: PMC11029593 DOI: 10.1007/s00262-015-1762-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 09/27/2015] [Indexed: 12/14/2022]
Abstract
The use of peptide vaccines, enhanced by adjuvants, has shown some efficacy in clinical trials. However, responses are often short-lived and rarely induce notable memory responses. The reason is that self-antigens have already been presented to the immune system as the tumor develops, leading to tolerance or some degree of host tumor cell destruction. To try to break tolerance against self-antigens, one of the methods employed has been to modify peptides at the anchor residues to enhance their ability to bind major histocompatibility complex molecules, extending their exposure to the T-cell receptor. These modified or analogue peptides have been investigated as stimulators of the immune system in patients with different cancers with variable but sometimes notable success. In this review we describe the background and recent developments in the use of analogue peptides for the immunotherapy of acute myeloid leukemia describing knowledge useful for the application of analogue peptide treatments for other malignancies.
Collapse
Affiliation(s)
- Susanne Hofmann
- Third Clinic for Internal Medicine, University of Ulm, Ulm, Germany
| | - Andrew Mead
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, LU1 3JU, UK
| | - Aleksandrs Malinovskis
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, LU1 3JU, UK
| | - Nicola R Hardwick
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Department of Haematological Medicine, Guy's, King's & St. Thomas' School of Medicine, The Rayne Institute, King's College London, 123 Coldharbour Lane, London, UK
| | - Barbara-Ann Guinn
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, LU1 3JU, UK.
- Department of Haematological Medicine, Guy's, King's & St. Thomas' School of Medicine, The Rayne Institute, King's College London, 123 Coldharbour Lane, London, UK.
- Cancer Sciences Unit, Southampton University Hospitals Trust, University of Southampton, Southampton, UK.
| |
Collapse
|
5
|
In Vitro and In Vivo Antitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL against CD33 Acute Myeloid Leukemia. Adv Hematol 2012; 2012:683065. [PMID: 22272203 PMCID: PMC3261457 DOI: 10.1155/2012/683065] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 10/05/2011] [Indexed: 11/18/2022] Open
Abstract
Genetic engineering of T cells with chimeric T-cell receptors (CARs) is an attractive strategy to treat malignancies. It extends the range of antigens for adoptive T-cell immunotherapy, and major mechanisms of tumor escape are bypassed. With this strategy we redirected immune responses towards the CD33 antigen to target acute myeloid leukemia. To improve in vivo T-cell persistence, we modified human Epstein Barr Virus-(EBV-) specific cytotoxic T cells with an anti-CD33.CAR. Genetically modified T cells displayed EBV and HLA-unrestricted CD33 bispecificity in vitro. In addition, though showing a myeloablative activity, they did not irreversibly impair the clonogenic potential of normal CD34(+) hematopoietic progenitors. Moreover, after intravenous administration into CD33(+) human acute myeloid leukemia-bearing NOD-SCID mice, anti-CD33-EBV-specific T cells reached the tumor sites exerting antitumor activity in vivo. In conclusion, targeting CD33 by CAR-modified EBV-specific T cells may provide additional therapeutic benefit to AML patients as compared to conventional chemotherapy or transplantation regimens alone.
Collapse
|
6
|
CD200 expression suppresses natural killer cell function and directly inhibits patient anti-tumor response in acute myeloid leukemia. Leukemia 2011; 25:792-9. [PMID: 21274000 DOI: 10.1038/leu.2011.1] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Upregulation of the immunosuppressive cell surface glycoprotein, CD200, is a common feature of acute myeloid leukemia (AML) and is associated with poor patient outcome. We investigated whether CD200 overexpression on AML cells could specifically compromise patient natural killer (NK) cell anti-tumor responses. We found that CD200(hi) patients showed a 50% reduction in the frequency of activated NK cells (CD56(dim)CD16(+)) compared with CD200(lo) patients. Additionally, NK receptor expression (NKp44 and NKp46) on these cells was also significantly downregulated in CD200(hi) patients. To assess whether NK cell activity was directly influenced by CD200 expression, we examined the effect of ectopic expression of CD200. These assays revealed that both NK cell cytolytic activity and interferon-γ response were significantly reduced toward CD200(+) leukemic targets and that these targets showed increased survival compared with CD200(-) cells. Similarly, NK cells isolated from AML patients were less functionally active toward CD200(hi) autologous blasts from both cytolytic and immunoregulatory perspectives. Finally, blocking CD200 alone was sufficient to recover a significant proportion of NK cell cytolytic activity. Together, these findings provide the first evidence that CD200 has a direct and significant suppressive influence on NK cell activity in AML patients and may contribute to the increased relapse rate in CD200(+) patients.
Collapse
|
7
|
Cen D, Hu G, Zhou Y, Yang L, Chen S, Schmidt CA, Li Y. Enhancement of specific cellular immune response induced by DNA vaccines encoding PML-RARalpha and hIL-2 genes. ACTA ACUST UNITED AC 2010; 15:88-95. [PMID: 20423569 DOI: 10.1179/102453310x125833470096589073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A DNA vaccine encoding PML-RAR alpha fusion gene is thought to be a promising approach for acute promyelocytic leukemia patients to enhance immune responses after attaining complete remission. In this study, we sought to enhance cellular immunity by coexpressing human interleukin (hIL)-2 genes. Successfully constructed plasmids PML-RAR alpha-hIL-2-pIRES, PML-RAR alpha-pIRES and hIL-2-pIRES were delivered intramuscularly in BALB/C mice at 14-day intervals for three cycles. The cellular immune responses with respect to the specific cytotoxicity of spleen cells; interferon-gamma secretion in sera, and the T-cell receptor rearrangement excision circles of thymocyte were significantly increased from PML-RARalpha-hIL-2-pIRES immunized mice. Our results indicate that a DNA vaccine with PML fusion gene segment and hIL-2 together might elicit increased cellular immune responses in mice.
Collapse
Affiliation(s)
- Dongzhi Cen
- Institute of Hematology, Medical College, Jinan University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
8
|
Depletion of endogenous tumor-associated regulatory T cells improves the efficacy of adoptive cytotoxic T-cell immunotherapy in murine acute myeloid leukemia. Blood 2009; 114:3793-802. [PMID: 19724059 DOI: 10.1182/blood-2009-03-208181] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumor-induced immune suppression can permit tumor cells to escape host immune resistance. To elucidate host factors contributing to the poor response of adoptively transferred tumor-reactive cytotoxic T lymphocytes (CTLs), we used a systemic model of murine acute myeloid leukemia (AML). AML progression resulted in a progressive regulatory T-cell (Treg) accumulation in disease sites. The adoptive transfer of in vitro-generated, potently lytic anti-AML-reactive CTLs failed to reduce disease burden or extend survival. Compared with non-AML-bearing hosts, transferred CTLs had reduced proliferation in AML sites of metastases. Treg depletion by a brief course of interleukin-2 diphtheria toxin (IL-2DT) transiently reduced AML disease burden but did not permit long-term survival. In contrast, IL-2DT prevented anti-AML CTL hypoproliferation, increased the number of transferred CTLs at AML disease sites, reduced AML tumor burden, and resulted in long-term survivors that sustained an anti-AML memory response. These data demonstrated that Tregs present at AML disease sites suppress adoptively transferred CTL proliferation, limiting their in vivo expansion, and Treg depletion before CTL transfer can result in therapeutic efficacy in settings of substantial pre-existing tumor burden in which antitumor reactive CTL infusion alone has proven ineffective.
Collapse
|
9
|
Cheuk ATC, Wells JW, Chan L, Westwood NB, Berger SA, Yagita H, Okumura K, Farzaneh F, Mufti GJ, Guinn BA. Anti-tumor immunity in a model of acute myeloid leukemia. Leuk Lymphoma 2009; 50:447-54. [PMID: 19197726 DOI: 10.1080/10428190802653776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Whole-cell vaccines allow the induction of anti-tumor immune responses without the need to define tumor antigens. We wished to directly compare, for the first time, the capacity of B7-1, B7-2 and 4-1BB ligand (4-1BBL) costimulatory molecules to convert murine and human acute myeloid leukemia (AML) cells into whole vaccines. 32Dc-kit is a murine myeloid cell line, which develops an AML-like disease over a protracted period, emulating human AML disease development. 32Dc-kit cells were modified to express elevated levels of B7-1, B7-2 or 4-1BBL, and each led to tumor rejection, although only mice injected with 32Dc-kit/B7-2 cells were able to reject subsequent parental tumor cell challenge. T-cell deficient nude mice were able to reject the 32Dc-kit variants, but they could not reject parental cell challenge; however, we found no evidence of cytotoxic T lymphocyte or natural killer (NK) activity ex vivo suggesting that tumor cell killing was mediated by an immune response that could not be recapitulated using purified NK or T cells as lone effectors. In human allogeneic mixed lymphocyte reactions (MLRs), we found no single costimulatory molecule was more effective, suggesting that the induction of a universal anti-tumor response will require a combination of costimulatory molecules.
Collapse
Affiliation(s)
- Adam T C Cheuk
- Department of Haematological Medicine, Allergy & Respiratory Science, MRC/Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London School of Medicine, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Gesundheit B, Shapira MY, Resnick IB, Amar A, Kristt D, Dray L, Budowski E, Or R. Successful cell-mediated cytokine-activated immunotherapy for relapsed acute myeloid leukemia after hematopoietic stem cell transplantation. Am J Hematol 2009; 84:188-90. [PMID: 19105234 DOI: 10.1002/ajh.21346] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Acute myeloid leukemia (AML) is an extremely aggressive disease with a high relapse rate even after allogeneic hematopoietic stem cell transplantation (HSCT). We report the successful outcome of cell-mediated cytokine-activated immunotherapy in a high-risk pediatric AML patient who relapsed shortly after allogeneic HSCT. Donor lymphocyte infusion along with interferon induced a graft-versus-leukemia effect, presenting as a reversible episode of graft-versus-host disease, which led to stable complete donor chimerism and total eradication of AML for over 24 months, at the time of this report. The curative potential of immunotherapy in hematological malignancies is discussed.
Collapse
Affiliation(s)
- Benjamin Gesundheit
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah University Hospital, Jerusalem, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Greiner J, Bullinger L, Guinn BA, Döhner H, Schmitt M. Leukemia-associated antigens are critical for the proliferation of acute myeloid leukemia cells. Clin Cancer Res 2008; 14:7161-6. [PMID: 19010831 DOI: 10.1158/1078-0432.ccr-08-1102] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. With intensive induction therapy, most patients younger than 60 years achieve complete remission. However, even if these younger patients were treated intensively, more than 50% will relapse. Clinical results of patients older than 60 years are more unfavorable. Therefore, in all patients with AML, the overall survival is still low. In the past decade, several leukemia-associated antigens (LAA) have been identified in patients with acute myeloid leukemia. BAGE, BCL-2, OFA-iLRP, FLT3-ITD, G250, hTERT, PRAME, proteinase 3, RHAMM, survivin, and WT-1 are all LAAs that have been shown to induce CD8+ T-cell recognition and for some antigens also humoral immune responses. Interestingly, most of these LAAs are linked to cell cycle or proliferation. This article discusses the balance between LAA-driven leukemia cell expansion and the elimination of these cells through attacks on LAAs by the immune system. Current knowledge of the function and CD8+ T-cell recognition of LAAs is reviewed and an outlook is given on how to improve T-cell responses to LAAs in acute myeloid leukemia cells.
Collapse
Affiliation(s)
- Jochen Greiner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany.
| | | | | | | | | |
Collapse
|
12
|
Krause DS, Van Etten RA. Right on target: eradicating leukemic stem cells. Trends Mol Med 2007; 13:470-81. [PMID: 17981087 PMCID: PMC4344828 DOI: 10.1016/j.molmed.2007.09.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 09/04/2007] [Accepted: 09/07/2007] [Indexed: 12/18/2022]
Abstract
Less than a third of adults with acute myeloid leukemia (AML) are cured by current treatments, emphasizing the need for new approaches to therapy. The discovery over a decade ago that myeloid leukemias originate from rare stem-like cells that can transfer the disease to immunodeficient mice suggested that these 'leukemia stem cells' (LSCs) are responsible for relapse of leukemia following conventional or targeted cancer therapy and that eradication of LSCs might be necessary to cure the disease permanently. Several recent studies have provided insight into the signaling pathways underlying the LSC phenotype and have also described approaches to eliminate LSCs with antibodies. Here, we review recent advances in LSC research and discuss novel therapeutic strategies to specifically target LSCs.
Collapse
Affiliation(s)
- Daniela S. Krause
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Richard A. Van Etten
- Molecular Oncology Research Institute and Division of Hematology/Oncology, Tufts-New England Medical Center, Boston, MA 02111, USA
| |
Collapse
|