1
|
Eissa MM, Salem AE, El Skhawy N. Parasites revive hope for cancer therapy. Eur J Med Res 2024; 29:489. [PMID: 39367471 PMCID: PMC11453045 DOI: 10.1186/s40001-024-02057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
Parasites have attained a life-long stigma of being detrimental organisms with deleterious outcomes. Yet, recently, a creditable twist was verified that can dramatically change our perception of those parasites from being a source of misery to millions of people to a useful anti-cancerous tool. Various parasites have shown promise to combat cancer in different experimental models, including colorectal, lung, and breast cancers, among others. Helminths and protozoan parasites, as well as their derivatives such as Echinococcus granulosus protein KI-1, Toxoplasma gondii GRA15II, and Trypanosoma cruzi calreticulin, have demonstrated the ability to inhibit tumor growth, angiogenesis, and metastasis. This article provides an overview of the literature on various cancer types that have shown promising responses to parasite therapy in both in vitro and in vivo animal studies. Parasites have shown anti-neoplastic activity through a variety of mechanisms that collectively contribute to their anti-cancer properties. These include immunomodulation, inhibition of angiogenesis, and molecular mimicry with cancer cells. This review article sheds light on this intriguing emerging field and emphasizes the value of collaborative multidisciplinary research projects with funding agencies and pharmaceutical companies. Thus, these strategies would secure continuous exploration of this new avenue and accelerate the advancement of cancer therapy research. Although experimental studies are heavily conducted by leaps and bounds, further steps are definitely lagging. Upgrading research from the experimental level to the clinical trial would be a wise progression toward efficient exploitation of the anti-neoplastic capabilities of parasites, ultimately saving countless lives.
Collapse
Affiliation(s)
- Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Ahmed Ebada Salem
- Department of Radiology and Nuclear Medicine, School of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 48123, USA
| | - Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Xie Y, Wang J, Wang Y, Wen Y, Pu Y, Wang B. Parasite-enhanced immunotherapy: transforming the "cold" tumors to "hot" battlefields. Cell Commun Signal 2024; 22:448. [PMID: 39327550 PMCID: PMC11426008 DOI: 10.1186/s12964-024-01822-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/08/2024] [Indexed: 09/28/2024] Open
Abstract
Immunotherapy has emerged as a highly effective treatment for various tumors. However, the variable response rates associated with current immunotherapies often restrict their beneficial impact on a subset of patients. Therefore, more effective treatment approaches that can broaden the scope of therapeutic benefits to a larger patient population are urgently needed. Studies have shown that some parasites and their products, for example, Plasmodium, Toxoplasma, Trypanosoma, and Echinococcus, can effectively transform "cold" tumors into "hot" battlefields and reshape the tumor microenvironment, thereby stimulating innate and adaptive antitumor immune responses. These parasitic infections not only achieve the functional reversal of innate immune cells, such as neutrophils, macrophages, myeloid-derived suppressor cells, regulatory T cells, and dendritic cells, in tumors but also successfully activate CD4+/CD8+ T cells and even B cells to produce antibodies, ultimately resulting in an antitumor-specific immune response and antibody-dependent cellular cytotoxicity. Animal studies have confirmed these findings. This review discusses the abovementioned content and the challenges faced in the future clinical application of antitumor treatment strategies based on parasitic infections. With the potential of these parasites and their byproducts to function as anticancer agents, we anticipate that further investigations in this field could yield significant advancements in cancer treatment.
Collapse
Affiliation(s)
- Yujun Xie
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Jinyan Wang
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yafei Wang
- Faculty of Arts and Science, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Yalin Wen
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yanping Pu
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Benfan Wang
- Laboratory of Tumor Immunobiology, Department of Public Health and Pathogen Biology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
- Institute of Surgery, The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China.
| |
Collapse
|
3
|
Sadr S, Borji H. Echinococcus granulosus as a Promising Therapeutic Agent against Triplenegative Breast Cancer. CURRENT CANCER THERAPY REVIEWS 2023; 19:292-297. [DOI: 10.2174/1573394719666230427094247] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/06/2022] [Accepted: 01/19/2023] [Indexed: 08/19/2024]
Abstract
Abstract:Breast cancer is a major cause of cancer deaths in women, with approximately 1.2 million new cases per year. Current treatment options for breast cancer include surgery, radiation, hormone therapy, and chemotherapy. However, the non-selective cytotoxicity of chemotherapeutic agents often leads to severe side effects, while drug resistance can worsen patient outcomes. Therefore, the development of more effective and less toxic anticancer drugs is a critical need. This study aimed to review the literature on Echinococcus granulosus antigens with anticancer potential against triple-negative breast cancer. Recent studies have suggested that certain parasite antigens may have potential anticancer effects. Specifically, research has shown that echinococcosis, a disease caused by the parasitic cestode Echinococcus granulosus, may have a protective effect against cancer. These findings offer new insights into the potential use of E. granulosus antigens in the development of novel cancer therapies and tumor cell vaccines. The findings of recent studies suggested that E. granulosus antigens may have the potential to be used in effective and less toxic cancer treatments. However, further research is needed to fully understand the mechanisms behind the anticancer effects of these antigens and develop new cancer therapies and vaccines
Collapse
Affiliation(s)
- Soheil Sadr
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
4
|
Asouli A, Sadr S, Mohebalian H, Borji H. Anti-Tumor Effect of Protoscolex Hydatid Cyst Somatic Antigen on Inhibition Cell Growth of K562. Acta Parasitol 2023:10.1007/s11686-023-00680-3. [PMID: 36991291 DOI: 10.1007/s11686-023-00680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND AND OBJECTIVE Today, cancer is one of the most important causes of death in the world, and so far, many treatment methods have been used in this field. Immunotherapy is considered one of the newest developments in this science, and it is still being investigated in some forms in different cancers and with a variety of antigens as well. One of the subsets of cancer immunotherapy is its treatment using parasitic antigens. The present study evaluated the effect of using somatic antigens of protoscoleces of Echinococcus granulosus on K562 cancer cells. METHODS In this study, hydatid cysts' protoscolex antigens were extracted, purified, and added to K562 cancer cells at three concentrations (0.1, 1, and 2 mg/ml) and on three times (24, 48, and 72 h). The number of apoptotic cells was compared to the control flask. The antigen concentration of 2 mg/ml was used as a control sample to investigate its cytotoxic effect on the growth of healthy HFF3 cells. Annexin V and PI tests were also performed to differentiate apoptosis from necrosis. RESULTS In flasks treated with hydatid cyst protoscolex antigen, all three concentrations significantly reduced the growth of cancer cells compared with the control flask, and concentration 2 of crude antigen significantly caused the death of cancer cells. Furthermore, more cancer cells underwent apoptosis by increasing the time of exposure to the antigen. On the other hand, flow cytometry results also showed that the amount of apoptosis has increased compared to the control group. In fact, Protoscolex hydatid cyst somatic antigens induce programmed cell death in K562 cancer cells while not having a cytotoxic effect on normal cells. CONCLUSION Therefore, it is suggested to do more research on the anti-cancer and therapeutic properties of the antigens of this parasite.
Collapse
Affiliation(s)
- Atefe Asouli
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran
| | - Soheil Sadr
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hadi Mohebalian
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 9177948974, Mashhad, Iran.
| |
Collapse
|
5
|
Porras-Silesky C, Mejías-Alpízar MJ, Mora J, Baneth G, Rojas A. Spirocerca lupi Proteomics and Its Role in Cancer Development: An Overview of Spirocercosis-Induced Sarcomas and Revision of Helminth-Induced Carcinomas. Pathogens 2021; 10:pathogens10020124. [PMID: 33530324 PMCID: PMC7911836 DOI: 10.3390/pathogens10020124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
Spirocerca lupi is a parasitic nematode of canids that induces a myriad of clinical manifestations in its host and, in 25% of infections, leads to the formation of sarcomas. The description of the protein composition of the excretory and secretory products (Sl-ESP) of S. lupi has shed light on its possible interactions with the host environment, including migration within the host and mechanisms of immunomodulation. Despite this, the process by which S. lupi induces cancer in the dog remains poorly understood, and some hypotheses have arisen regarding these possible mechanisms. In this review, we discuss the role of specific ESP from the carcinogenic helminths Clonorchis sinensis, Opisthorchis viverrini and Schistosoma haematobium in inducing chronic inflammation and cancer in their host’s tissues. The parasitic worms Taenia solium, Echinococcus granulosus, Heterakis gallinarum, Trichuris muris and Strongyloides stercoralis, which have less-characterized mechanisms of cancer induction, are also analyzed. Based on the pathological findings in spirocercosis and the mechanisms by which other parasitic helminths induce cancer, we propose that the sustained inflammatory response in the dog´s tissues produced in response to the release of Sl-ESP homologous to those of other carcinogenic worms may lead to the malignant process in infected dogs.
Collapse
Affiliation(s)
- Catalina Porras-Silesky
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - María José Mejías-Alpízar
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - Javier Mora
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - Gad Baneth
- Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Alicia Rojas
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
- Correspondence: ; Tel.: +506-2511-8644
| |
Collapse
|
6
|
Guan W, Zhang X, Wang X, Lu S, Yin J, Zhang J. Employing Parasite Against Cancer: A Lesson From the Canine Tapeworm Echinococcus Granulocus. Front Pharmacol 2019; 10:1137. [PMID: 31607934 PMCID: PMC6774290 DOI: 10.3389/fphar.2019.01137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/03/2019] [Indexed: 01/03/2023] Open
Abstract
Cystic echinococcosis (CE), a devastating zoonotic condition caused by the tapeworm Echinococcus granulosus, remain a significant public health problem worldwide. However, after a negative correlation between solid tumor and CE has been incidentally discovered, accumulating evidence have suggested that this parasite may induce anticancer effect through activating host immune response and secreting molecules with anticancer potential, which may provide some new understanding for immunotherapy. This article will review the evidence supporting the anticancer effect of E. granulosus and its underlying mechanisms and discuss the possible implications in immunotherapy.
Collapse
Affiliation(s)
- Wang Guan
- Department of Cancer Prevention and Treatment, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoqin Zhang
- Intensive Care Unit, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Wang
- Department of Cancer Prevention and Treatment, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shun Lu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Yin
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinxin Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Mohamadi F, Shakibapour M, Sharafi SM, Reza AA, Tolouei S, Darani HY. Anti- Toxoplasma gondii antibodies attach to mouse cancer cell lines but not normal mouse lymphocytes. Biomed Rep 2019; 10:183-188. [PMID: 30906547 DOI: 10.3892/br.2019.1186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 01/04/2019] [Indexed: 01/03/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is prevalent intracellular parasite and a cause of worldwide infection in the human population. An inhibitory effect of this parasite on cancer growth has been demonstrated in cell culture and animal models. To determine whether the anticancer activities of T. gondii are associated with host immune response, in the current study the reactivity of anti-T. gondii antiserum with the surface of cancer cell lines was investigated. Anti-T. gondii antibodies were raised in rabbit and the reaction of this antiserum in comparison with other anti-parasite antisera (anti-T. vaginalis, anti-hydatid cyst fluid, anti-protoscolices antigens) with mouse melanoma or breast cancer cells lines was investigated using flow cytometry. Anti-T. gondii antiserum reacted markedly with the surface of mouse melanoma and breast cancer cells, and less so with the normal mouse spleen lymphocytes. Meanwhile, the other anti-parasite antisera did not react strongly with the surface of cancer cells compared with normal mouse spleen lymphocytes. In summary, it has been demonstrated herein that anti-T. gondii antiserum may selectively react with the surface of mouse cancer cells but not with normal mouse spleen lymphocytes. Therefore, further study on anti-Toxoplasma antibodies may be useful for directing the application of selective drug delivery in cancer treatment.
Collapse
Affiliation(s)
- Fereshteh Mohamadi
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Mahshid Shakibapour
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Seyedeh Maryam Sharafi
- Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Ali Andalib Reza
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Sepideh Tolouei
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| | - Hossein Yousofi Darani
- Department of Medical Parasitology and Mycology, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran.,Cancer Prevention Research Centre, Isfahan University of Medical Sciences, Isfahan 8179498861, Iran
| |
Collapse
|
8
|
Intrapulmonary teratoma masquerading as hydatid cyst of the lung. Indian J Thorac Cardiovasc Surg 2019; 35:71-73. [PMID: 33060975 DOI: 10.1007/s12055-018-0701-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/06/2018] [Accepted: 07/13/2018] [Indexed: 10/28/2022] Open
Abstract
Intrapulmonary teratoma (IPT) is a rare mediastinal teratoma. IPT arise because of abnormal migration of pluripotent stem cells along the developing lung bud. Patients present with chest pain, hemoptysis, and trichoptysis. These tumors have the potential to rupture into adjacent pleura, pericardium, or tracheobronchial tree. The differential diagnosis includes hydatid cyst, fungal ball, lung abscess, and lung parenchymal tumor. We report a case of IPT, which was initially diagnosed as hydatid cyst of the lung due to positive hydatid serology and correlating radiological features. The role of cross-reaction of hydatid antigens with sera of cancer patients needs to be evaluated. Surgical resection of the tumor is advised to avoid complications.
Collapse
|
9
|
Gao XY, Zhang GH, Huang L. Modulation of human melanoma cell proliferation and apoptosis by hydatid cyst fluid of Echinococcus granulosus. Onco Targets Ther 2018; 11:1447-1456. [PMID: 29588599 PMCID: PMC5858823 DOI: 10.2147/ott.s146300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective The objective of this paper was to assess the effects of hydatid cyst fluid (HCF) of Echinococcus granulosus on melanoma A375 cell proliferation and apoptosis. Methods A375 cells were classified into five groups by in vitro culture: normal group, control group, 10% HCF group, 20% HCF group and 30% HCF group. Trypan blue staining method was employed to detect the toxicity of HCF. Effects of different concentrations of HCF on melanoma A375 cell proliferation at different time points were evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Flow cytometry and propidium iodide (PI) staining were used to detect cell cycle, and Annexin-V/PI double staining method was used to determine A375 cell apoptotic rate. Western blotting was applied to detect the expression of phosphorylated extracellular regulated protein kinases, proliferating cell nuclear antigen (PCNA), cell-cycle-related proteins (cyclin A, cyclin B1, cyclin D1 and cyclin E) and apoptosis-related proteins (Bcl-2, Bax and caspase-3). Results HCF with a high concentration was considered as atoxic to A375 cells. HCF promoted A375 cell proliferation, and the effects got stronger with an increase in concentrations but was retarded after reaching a certain range of concentrations. HCF increased phosphorylation level and expression of extracellular regulated protein kinase, as well as PCNA expression. HCF also promoted the transferring progression of A375 cells from the G0/G1 phase to the S phase to increase the cell number in S phase and increased the expression of cyclin A, cyclin D1 and cyclin E. HCF increased the expression of procaspase-3 (the precursor of apoptosis-related protein caspase-3) and antiapoptotic protein-Bcl-2, and decreased the expression of proapoptotic factor Bax, thereby inhibiting cell apoptosis. Conclusion As a result, this study confirmed that HCF promotes proliferation and inhibits apoptosis of melanoma A375 cells.
Collapse
Affiliation(s)
- Xiang-Yang Gao
- Department of Laboratory Medicine, Pu'er People's Hospital, Pu'er
| | - Guang-Hui Zhang
- Department of Clinical Laboratory, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai
| | - Li Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai, China
| |
Collapse
|
10
|
Ranasinghe SL, McManus DP. Echinococcus granulosus: Cure for Cancer Revisited. Front Med (Lausanne) 2018; 5:60. [PMID: 29594121 PMCID: PMC5857532 DOI: 10.3389/fmed.2018.00060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
Whereas a number of parasites are well recognized risk factors for a number of different cancers in mammalian hosts, there is limited information on the ability of parasitic organisms to induce anticancer effects. There are conflicting reports that echinococcosis, caused by the canine tapeworm Echinococcus granulosus, can decrease or increase cancer risk. This review considers both indirect anticancer effects as the result of adaptive immunity generated against certain echinococcal antigens and the direct effect of molecules released by E. granulosus whose activity directly inhibits cancer cell migration and growth. In conclusion, E. granulosus probably secretes molecules that can be developed as anticancer therapeutics in future.
Collapse
Affiliation(s)
- Shiwanthi L Ranasinghe
- Molecular Parasitology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Donald P McManus
- Molecular Parasitology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|