相关文章推荐
文雅的炒饭  ·  SKStoreReviewControlle ...·  3 月前    · 
英俊的双杠  ·  Idea JSTL 500 - 知乎·  1 年前    · 
开朗的茄子  ·  jQuery UI 实例 – ...·  1 年前    · 
The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely. As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health. Learn more about our disclaimer. Phase 1Refractory malignant solid neoplasmTCR-T cells, cyclophosphamide and fludarabine {"type":"clinical-trial","attrs":{"text":"NCT04809766","term_id":"NCT04809766"}} NCT04809766 Phase 1Solid tumorsanti-MSLN CAR-T cells {"type":"clinical-trial","attrs":{"text":"NCT03545815","term_id":"NCT03545815"}} NCT03545815 [ 64 ]Phase 1Lung adenocarcinoma, ovarian cancer,
peritoneal carcinomaanti-MSLN CAR-T cells {"type":"clinical-trial","attrs":{"text":"NCT03054298","term_id":"NCT03054298"}} NCT03054298 [ 65 ]Phase 1Malignant pleural mesotheliomaanti-MSLN CAR-T cells and
cyclophosphamide {"type":"clinical-trial","attrs":{"text":"NCT04577326","term_id":"NCT04577326"}} NCT04577326 Phase 1MSLN+ neoplasms, epithelioid mesothelioma, cholangiocarcinoma, pancreatic adenocarcinomaLMB-100 and Tofacitinib {"type":"clinical-trial","attrs":{"text":"NCT04034238","term_id":"NCT04034238"}} NCT04034238 [ 66 ]Phase 1MesotheliomaLMB-100 and Ipilimumab {"type":"clinical-trial","attrs":{"text":"NCT04840615","term_id":"NCT04840615"}} NCT04840615 Phase 1Advanced recurrent epithelioid mesothelioma, serous ovarian cancer, metastatic or locally advanced pancreatic ductal adenocarcinomaBAY2287411 (MSLN-TTC) {"type":"clinical-trial","attrs":{"text":"NCT03507452","term_id":"NCT03507452"}} NCT03507452 [ 67 ]Phase 1, 2Advanced MSLN+ tumors HPN536 {"type":"clinical-trial","attrs":{"text":"NCT03872206","term_id":"NCT03872206"}} NCT03872206 Phase 1, 2Pancreatic adenocarcinomaAnetumab ravtansine, gemcitabine hydrochloride, Ipilimumab and Nivolumab {"type":"clinical-trial","attrs":{"text":"NCT03816358","term_id":"NCT03816358"}} NCT03816358 Phase 2Fallopian tube endometrioid adenocarcinoma, high grade fallopian tube and ovarian serous adenocarcinomaAnetumab ravtansine,
Bevacizumab and paclitaxel {"type":"clinical-trial","attrs":{"text":"NCT03587311","term_id":"NCT03587311"}} NCT03587311 Phase 2Non-small cell lung cancerLMB-100 and Pembrolizumab {"type":"clinical-trial","attrs":{"text":"NCT04027946","term_id":"NCT04027946"}} NCT04027946 CompletedMalignant. MesotheliomaSS1P, pentostatin, and
cyclophosphamide {"type":"clinical-trial","attrs":{"text":"NCT01362790","term_id":"NCT01362790"}} NCT01362790 CompletedPancreatic cancerGVAX and CRS-207 {"type":"clinical-trial","attrs":{"text":"NCT02243371","term_id":"NCT02243371"}} NCT02243371 [ 68 ]

The response to immunotherapy agents is often restricted by the interactions with immune checkpoint proteins. Programmed cell death protein 1 (PD-1) is one such extracellular protein expressed on activated T cells that helps modulate the T cell response. Programmed cell death ligand 1 (PD-L1) is expressed on antigen-presenting cells (APCs) and the binding of PD-1 and PD-L1 is considered coinhibitory as it can reduce T cell activity through reduction in cytokine secretion [ 69 ]. The expression of PD-L1 on tumor cells limits the efficacy of immune cells, as these cells do not recognize the tumor cell as their ideal target. Therefore, immune checkpoint blockade therapies are used in combination with immunotherapeutics to overcome the immune evasive response.

6.1. Immunotoxins and Monoclonal Antibodies

SS1P is an anti-mesothelin immunotoxin, an antibody-based therapy with a bacterial toxin payload. This recombinant protein consists of a murine Fv fragment linked to a Pseudomonas exotoxin A payload [ 70 ]. Although the toxin was well tolerated and exhibited significant anti-tumor activity [ 59 ], the efficacy was limited by the development of neutralizing antibodies. This limitation was overcome by combining SS1P with chemotherapy agents that suppress the host immune system [ 71 ]. LMB-100 is a second-generation immunotoxin comprising a humanized anti-mesothelin fragment and a modified Pseudomonas exotoxin A payload which was designed to be less immunogenic [ 72 ]. LMB-100 has been similarly limited by the development of anti-drug antibodies [ 73 , 74 ]. In mesothelioma, the combination of LMB100 and anti-PD-1 antibody therapy has enhanced efficacy comparative to solo therapies [ 75 ]. Both modalities continue to be studied in combination with chemotherapy in mesothelin-expressing solid tumors.

Amatuximab (originally called MORAb-009) is a monoclonal antibody which targets mesothelin, thereby disrupting cell adhesion and initiating antibody-dependent cytotoxicity [ 76 ]. The anti-tumor effect and maximum tolerated dose have been established in completed phase I and II clinical trials in combination with chemotherapy [ 77 , 78 ]. In vitro studies indicate that amatuximab inhibits mesothelin interaction with CA125/MUC16 and such a blockage reduces the cancer’s ability to metastasize and invade into other tissues. Additionally, amatuximab treatment downregulated cancer stem cell markers, such as CD44, c-MET, and ALDH1 in pancreatic cancer cells [ 79 ]. Amatuximab treatment increased sensitivity to gemcitabine and reduced the expression of c-Met and AKT in the liver, accompanied by decreased rates of pancreatic cancer cell metastasis [ 79 , 80 ].

6.2. Vaccines

GVAX is a granulocyte–macrophage colony-stimulating factor (GM-CSF) tumor cell vaccine which expresses multiple antigens and induces anti-tumor immune responses by cross-priming mechanisms to recruit antigen-presenting cells [ 81 ]. Mesothelin was identified as a target of T cells in patients treated with GVAX, initiating the development of a Listeria monocytogenes vaccine (CRS-207), which expresses tumor-associated antigen mesothelin. CRS-207 secretes mesothelin in the cytosol of infected antigen-presenting cells, which is processed and presented by major histocompatibility complex (MHC), thereby stimulating an immune response [ 82 , 83 ]. A phase II study using CRS-207 in combination with GVAX and conventional chemotherapy for patients with previously treated metastatic pancreatic adenocarcinoma improved overall survival [ 84 ]; however, a follow-up phase IIb study showed similar survival as chemotherapy [ 85 ]. A phase I study of CRS-207 in addition to conventional chemotherapy for the treatment of patients with malignant pleural mesothelioma found that it was well tolerated, with few adverse events. They reported reduced tumor sizes and an improvement in survival, including one complete response [ 86 ].

6.3. Antibody–Drug Conjugates

Anetumab ravtansine is an antibody–drug conjugate (ADC) comprised of a humanized anti-mesothelin antibody conjugated to DM4 (a maytansinoid tubulin inhibitor), which showed potent killing of tumor cells expressing mesothelin [ 87 ]. The drug binds specifically to mesothelin-expressing tumor cells and releases its cytotoxic payload following internalization. Phase I trials in patients with advanced solid tumors suggested that anetumab ravtansine can be safely given to patients and supported its anti-tumor efficacy [ 88 ]. Additional phase I and II trials are ongoing. Preclinical studies showed an improved anti-tumor effect when used in combination with standard chemotherapy in ovarian cancer models [ 89 ].

ADC DMOT4039A, a humanized anti-mesothelin monoclonal antibody conjugated to monomethyl auristatin E (MMAE), which has anti-mitotic effects, was found to be safe in a phase I clinical trial [ 90 ].

Another immunoconjugate, BMS-986148, conjugated to tubulysin, which disrupts microtubule assembly and induced apoptosis, was evaluated in solid tumors. Phase 1/2a studies of BMS-986148 treatment alone or in combination to nivolumab, a PD-1 inhibitor, was concluded to have manageable safety as well as clinical activities, warranting further clinical trials in combination with other checkpoint inhibitors [ 91 ].

6.4. Chimeric Antigen Receptor T Cells and T Cell Receptor Fusion Constructs

Chimeric Antigen Receptor (CAR) T cells are autologous patient T cells which are genetically modified to target a cancer-specific protein. When these cells bind to their target, they become activated, proliferate and mediate cytotoxic effects [ 92 ]. CAR-meso T cells consist of an anti-mesothelin scFv fused to TCRzeta signaling and costimulatory domains, allowing for specific binding to mesothelin expressing cells and subsequent cytotoxic response [ 93 , 94 ]. Preclinical studies of mesothelin targeting CAR-T cells have shown significant tumor reduction and are currently under clinical evaluation by many groups [ 93 , 95 ]. Phase I studies have shown that mesothelin targeting CAR-T cells are well tolerated with minimal on-target off-tumor effects but showed limited clinical activity [ 65 ]. There are many ongoing trials investigating mesothelin-directed CAR-T cells in solid tumors [ 96 ].

Most recently, novel T cell engineering platforms which target mesothelin have been investigated. T cell receptor fusion constructs (TRuCs) target tumor cells independent of MHC, resulting in increased T cell activation. Preclinical studies with mesothelin-directed TRuCs have shown robust anti-tumor activity, with faster rates of accumulation in mesothelin-expressing tumors, lower levels of cytokines, increased levels of chemokine receptors, and long-term functional persistence [ 97 ].

CAR-T Cell Alterations

CAR-T cells are conventionally designed against single antigens of interest; however, Tandem CAR-T cells targeting two antigens may be more effective and have higher anti-tumor effects than single antigen targeting CAR-Ts. These Tandem CAR-Ts allow for simple Boolean logic gates of “AND”, “OR”, or “NOT”, as they can recognize multiple antigens, and have been used in hematological malignancies, including acute lymphoblastic leukemia to simultaneously target CD19 and CD20 [ 98 , 99 ]. Tandem CAR-Ts targeting cancer-specific upregulated molecules mesothelin and folate receptor 1 (FOLR1) with secretory activity of IL-12 had higher infiltration and persistence comparative to anti-mesothelin CAR-Ts, and the secretion of IL-12 enhanced therapeutic effects and reduced tumor antigen escape in ovarian cancer [ 100 ]. These data suggest that the identification of multiple tumor-associated antigens would improve the results of CAR-T cells by applying Boolean logic for the recognition of multiple targets to reduce antigen escape.

The silencing of PD-1 on CAR-T cells with the use of shRNA resulted in increased efficacy and an enhanced anti-tumor effect on several mesothelin-expressing cancers [ 101 ]. The success of PD-1 silencing in CAR-T cell therapy opens the door to the gene silencing or activation of other genes in these cells to improve anti-tumor effects. Another antigen of interest with CAR-T cells is Tim3 (T cell immunoglobulin domain and mucin domain 3), which is an immune checkpoint receptor that helps regulate T cell response in the tumor microenvironment. Tim3 plays a role in immunosuppression and T cell death. Blocking Tim3 function reduced immunosuppression, reduced regulatory T cells, and increased IFN-γ production from T cells [ 102 , 103 ]. Blocking Tim3 expression on mesothelin CAR-T cells through shRNA had improved cytotoxicity effects, increased cytokine production, and higher proliferation capacity compared to their Tim3+ counterparts [ 104 ]. These two studies present the possibility of customizing CAR-T cells to overcome mechanisms that inhibit their function within the tumor microenvironment and will need further investigation to identify and silence other immunosuppressive targets.

Another factor that decreases the efficacy of CAR-T cells is the production of adenosine in the tumor microenvironment. The overproduction of adenosine from tumor cells facilitates immunosuppression through the binding of the adenosine 2a receptor (A2aR). The knockdown of A2aR through shRNA in CAR-T cells increased the anti-tumor ability of the CAR-T cells both in vivo and in vitro. In xenograft models, the A2aR-disrupted anti-mesothelin CAR-T cells decreased the tumor burden compared to unmodified T cells and anti-mesothelin CAR-T cells [ 105 ]. Expression of anti-mesothelin CAR-T cells with cell chemokine receptors CCR2b and CCR4 revealed enhanced migration of CAR-Ts in vitro and displayed high levels of cytotoxicity and increased levels of IL-2, IFN-γ, and TNF-α, revealing CCR2b as the superior chemokine receptor in NSCLC in vivo studies [ 106 ].

6.5. Chimeric Antigen Receptor Natural Killer Cells

Similar to CAR-T cells, natural killer (NK) cells can also express chimeric antigen receptors and are possibly more effective than CAR-T cells due to their increased ability to recognize and kill tumor cells to result in tumor-specific killing. In a study on ovarian cancer, CAR-NK cells were designed to recognize mesothelin, and were effective in eliminating mesothelin+ cancer cells through increased cytokine secretion compared to the parental NK cell treatment [ 107 ]. In an in vitro study using anti-mesothelin NK cells against gastric cancer, mesothelin-NK cells were more selective in killing mesothelin+ tumors and secreting cytokines comparative to transduced NK cells and were able to prolong survival rates of patient-derived xenograft (PDX) mice in vivo [ 108 ].

6.6. Bispecific T Cell-Engaging Molecules

Bispecific T cell engagers (BiTEs) are small fusion proteins consisting of two single chain variable fragments (scFvs): one which targets an effector cell (most commonly CD3 on T cells) and another which targets the tumor antigen. These bispecific antibodies redirect the cytotoxic effects of T cells toward specific tumor cells [ 109 ]. While the use of BiTEs has been most notable in hematologic malignancies and B cell acute leukemias in particular, they have been developed for a number of different tumor antigens. Research into the use of BiTEs which target mesothelin are currently underway in triple-negative breast cancer, pancreatic ductal adenocarcinoma, lung cancer, and other solid tumors [ 110 , 111 ].

One drawback to using BiTEs in vivo is their short half-life of a few hours, thereby requiring administration via intravenous infusions. To overcome this limitation, Suurs et al. designed an anti-mesothelin/anti-CD3 BiTE that also had an Fc-domain that increased the half-life and required only intermittent dosing of BiTEs. PET imaging in BALB/c mice revealed the tumor-tissue specific uptake of the BiTE molecule and a half-life of about 63 h [ 112 ]. A similar bispecific antibody in the IgG format with bivalency for mesothelin in addition to binding CD3 showed higher efficacy than a corresponding antibody with monovalent mesothelin binding capacity [ 113 ]. A trispecific T cell-activating construct, HPN536, has been used to treat mesothelin-expressing solid tumors. In addition to binding to CD3 and mesothelin, HPN536 binds serum albumin to increase the plasma half-life of the molecule. In in vivo studies of cynomolgus monkeys, HPN536 was well tolerated and exhibited mesothelin-dependent pharmacokinetics, which led to a phase I clinical trial ( {"type":"clinical-trial","attrs":{"text":"NCT03872206","term_id":"NCT03872206"}} NCT03872206 ) using HPN536 against solid tumors [ 114 ].

6.7. Targeted Alpha Therapies

Targeted alpha therapies (TAT) represent a new emerging class of targeted cancer therapies that use high-energy emissions from alpha particles to elicit permanent double-stranded breaks in DNA, ultimately resulting in cell death [ 115 ]. Targeted thorium-227 conjugates (TTC) represent a subtype of TATs that consist of a covalently attached 3,2-HOPO chelator to a specified antibody to ensure the delivery of thorium-227, an alpha particle emitter, to mesothelin-expressing cells. The specific mesothelin TTC, BAY 2287411 in mesothelioma, ovarian, and breast cancers, among others, showed anti-tumor potency both in vitro and in vivo in PDX models. The cellular response of BAY 2287411 included increased DNA double-stranded breaks, oxidative stress, and apoptotic markers. The results from this study support the transition of BAY 2287411 to a phase I clinical trial to treat ovarian and mesothelioma cancers [ 67 ]. A combination therapy of BAY 2287411 with DNA damage response inhibitors results in a synergistic effect by sensitizing the cells to DNA damage [ 116 ]. Targeted alpha therapies remain as one of the least investigated targeted therapeutic options when treating mesothelin-positive tumors.

6.8. Impact of Mesothelin Shedding on Mesothelin-Targeting Therapies

As previously noted, many GPI-anchored proteins undergo routine extracellular shedding, to which mesothelin is no exception. Shed mesothelin could decrease the efficacy of targeted therapies, resulting in an on-target, off-tumor response of the drugs interacting with soluble mesothelin. The most notable sheddase involved in mesothelin shedding is TACE/ADAM17; however, many members of the ADAM, MMP, and BACE protease families are able to shed mesothelin from the cell surface. Inhibiting the shedding of mesothelin with drugs that limit functionality of sheddases has shown promising results in vitro by increasing drug efficacy and cell surface mesothelin [ 16 , 61 ]. The identification of an epitope on the fragmented mesothelin that remains unshed on the surface of the membrane would be more effective in targeting mesothelin.

7. Role of Mesothelin in Hematological Malignancies

The overexpression of mesothelin has been well described in several solid tumors; however, there are limited data regarding its expression in hematologic malignancies. Mesothelin expression was lacking in all 442 tissue microarray specimens, representing different types of lymphoma. This study did not evaluate samples from other hematological malignancies [ 41 ]. In 2006, Steinbach et al. identified MSLN as one of the seven genes overexpressed in pediatric AML compared to normal bone marrow [ 117 ]. Further investigation detected mesothelin protein expression in a small number of primary pediatric AML samples [ 118 ]. Recently, massive sequencing efforts in pediatric AML under the direction of Children’s oncology group and NCI (TARGET initiative) identified mesothelin to be expressed in greater than a third (36%) of AML samples [ 11 ]. Mesothelin expression was quantified and subsequently confirmed using quantitative RT-PCR and flow cytometry. Data from TCGA and BEAT databases showed that only about 14% of adult AML samples expressed mesothelin. Among the various cytogenetic subtypes of pediatric AML, core binding factor AML (comprising inv(16), t(8;21)), and KMT2A rearranged AML comprised 49% and 40% of samples with mesothelin positivity, respectively.

How mesothelin expression is triggered in AML is not known. In pediatric AML patients, genomic characterization revealed that mesothelin expression is inversely correlated with MSLN promoter hypermethylation, consistent with previously described data from solid tumor studies. AML samples with high promoter methylation were characterized as low mesothelin expressers via flow cytometry [ 11 ].

Preclinical studies to evaluate the efficacy of mesothelin-targeted therapies such as immunotoxin, ADC, BiTEs, CAR-T cells, and CAR-NK cells in pediatric AML have been conducted ( Figure 3 ). An early study using recombinant anti-mesothelin immunotoxin SS1(dsFv)PE38 in vitro failed to produce a cytotoxic effect. The authors speculated that other mesothelin-targeted approaches could be more beneficial [ 118 ]. Accordingly, both in vitro and in vivo evaluations of ADC anteumab ravtansine in AML samples resulted in the selective killing of mesothelin-expressing cell lines. Mice xenografted with mesothelin+ samples survived longer when treated with the ADC compared to the isotype drug conjugate control and mesothelin-negative samples exposed to either treatment. Another MSLN-ADC, DGN462, bearing a DNA alkylating agent, also showed preclinical efficacy in pediatric AML [ 11 ]. The combination of the ADC treatment with traditional chemotherapy regimen of daunorubicin and cytarabine led to higher survival rates compared to any treatment alone, suggesting a synergistic effect [ 119 ]. A phase I clinical trial for anetumab ravtansine is currently under development by the Children’s Oncology Group for mesothelin+ pediatric AML patients in second relapse [ 120 ].

Mesothelin-targeted therapies in preclinical development for pediatric AML. Anetumab ravtansine is an antibody–drug conjugate (ADC) that is an scFv for mesothelin attached to DM4 (red star), a tubulin inhibitor, via a linker (yellow). Once bound to mesothelin, DM4 is internalized and binds to tubulin, which disrupts microtubule assembly and cell cycle is inhibited, which results in cell death. Chimeric antigen receptor (CAR) therapy uses a variety of host immune cells to elicit a tumor-targeted response using a construct designed to recognize a target antigen. In AML, CAR-NK cells (yellow), and CAR T cells (orange) have been investigated. Upon the recognition of the target antigen, NK and T cells activate and elicit a killing mechanism through the secretion of perforin and granzymes (blue), resulting in tumor cell lysis. Bispecific T cell-engaging molecules (BiTEs) work mechanistically by recruiting T cells to tumor cells by engaging with a tumor-associated antigen (shown in blue), while the other side of the molecule engages with a CD3 on a T cell (shown in green). Once bound, the T cell becomes activated and kills through the same mechanisms at the CAR cell lysis.

Mesothelin-targeting BiTEs were found to be highly effective at inducing complete remission in two PDX models of KMT2A rearranged AML. Because these studies were performed in immunodeficient mice, human T cells were injected to act as effector cells. BiTE molecules in the IgG format with a longer in vivo half-life were able to suppress the extramedullary tumor masses that are commonly observed in MV4;11 xenografted mice [ 63 ]. CAR-T cells targeting mesothelin were efficacious in controlling tumor burden by eradicating leukemia stem cells [ 61 ]. Most recently, work has been conducted using anti-mesothelin CAR NK-92 cells in AML samples. In vitro and in vivo studies revealed that these CAR NK-92 cells were effective at specifically targeting mesothelin+ AML samples while not killing mesothelin-negative samples [ 62 ]. These different modalities that target mesothelin provide effective targeted therapeutic options for mesothelin+ AML.

What is the role of mesothelin in AML? Of important clinical significance, in a review of patients with core binding factor AML, which traditionally confers a more favorable risk stratification, those with concurrent mesothelin expression had a statistically significant higher risk of relapse and worse disease-free survival [ 121 ]. Mesothelin expression was significantly associated with the presence of extramedullary disease, which may portend poor survival in AML patients [ 63 ]. Interestingly, mesothelin promoted leukemia growth and was also associated with extramedullary disease in AML xenograft models [ 122 ]. Mesothelin expression in leukemia stem cells suggests that similar to solid tumors, mesothelin may play a role in leukemia progression and relapse [ 61 ]. Further studies are necessary to shed light on the role of mesothelin in hematological malignancies.

In summary, recent findings of mesothelin overexpression in pediatric AML suggest this may be a useful target for therapy. Given the overall poor outcomes for children with AML, further studies to explore the use of mesothelin-directed immunotherapy are warranted. Pre-clinical studies utilizing anti-mesothelin CAR-T cells, BiTEs, and anetumab ravtansine against mesothelin-expressing AML exhibit tumor-specific killing of leukemia cells, and the ability to reduce leukemic burden in vivo. These studies of monotherapies as well as combination therapies with chemotherapy and/or immune checkpoint blockade inhibitors present a strong case for transition into clinical studies.

8. Conclusions

Mesothelin is a highly desirable target for tumor-specific immunotherapy given its overexpression in tumor cells and relative paucity in normal healthy cells. Currently, research seems to focus on the use of mesothelin to produce promising targeted therapy approaches, and to match the needs of the patient population and the tumor microenvironment specific to each cancer type. Many different targeted approaches allow for slight alterations that can improve and make the therapies more targeted in relation to difficulties, such as silencing immunosuppressive molecules. There still is a need, however, to identify the function of mesothelin, and other key characteristics of mesothelin, such as the unshed epitope to increase the therapeutic potential. While mesothelin has been considered a target for solid tumors, recent findings of mesothelin overexpression in AML point towards a further evaluation of the expression of this marker in other hematological malignancies. The absence of mesothelin on normal and healthy hematopoietic cells is of particular importance, as currently available immunotherapies for the treatment of AML are associated with hematopoietic toxicities.

Author Contributions

J.R.F. and D.H. wrote the manuscript. A.G., D.H., E.A.K., and S.P.B. conceptualized the work and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Leukemia Research Foundation of Delaware, Lisa Dean Moseley Foundation, and the Nemours Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

Footnotes

Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

1. Chang K., Pastan I., Willingham M.C. Isolation and characterization of a monoclonal antibody, K1, reactive with ovarian cancers and normal mesothelium. Int. J. Cancer. 1992; 50 :373–381. doi: 10.1002/ijc.2910500308. [ PubMed ] [ CrossRef ] [ Google Scholar ]
2. Chang K., Pastan I. Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers. Proc. Natl. Acad. Sci. USA. 1996; 93 :136–140. doi: 10.1073/pnas.93.1.136. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
3. Hassan R., Wu C., Brechbiel M.W., Margulies I., Kreitman R.J., Pastan I. 111Indium-labeled monoclonal antibody K1: Biodistribution study in nude mice bearing a human carcinoma xenograft expressing mesothelin. Int. J. Cancer. 1999; 80 :559–563. doi: 10.1002/(SICI)1097-0215(19990209)80:4<559::AID-IJC13>3.0.CO;2-Y. [ PubMed ] [ CrossRef ] [ Google Scholar ]
4. Lamberts L.E., Menke-van der Houven van Oordt C.W., ter Weele E.J., Bensch F., Smeenk M.M., Voortman J., Hoekstra O.S., Williams S.P., Fine B.M., Maslyar D., et al. ImmunoPET with Anti-Mesothelin Antibody in Patients with Pancreatic and Ovarian Cancer before Anti-Mesothelin Antibody-Drug Conjugate Treatment. Clin. Cancer Res. 2016; 22 :1642–1652. doi: 10.1158/1078-0432.CCR-15-1272. [ PubMed ] [ CrossRef ] [ Google Scholar ]
5. Hilliard T.S. The Impact of Mesothelin in the Ovarian Cancer Tumor Microenvironment. Cancers. 2018; 10 :277. doi: 10.3390/cancers10090277. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
6. Montemagno C., Cassim S., Pouyssegur J., Broisat A., Pages G. From Malignant Progression to Therapeutic Targeting: Current Insights of Mesothelin in Pancreatic Ductal Adenocarcinoma. Int. J. Mol. Sci. 2020; 21 :4067. doi: 10.3390/ijms21114067. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
7. Hagerty B.L., Pegna G.J., Xu J., Tai C.H., Alewine C. Mesothelin-Targeted Recombinant Immunotoxins for Solid Tumors. Biomolecules. 2020; 10 :973. doi: 10.3390/biom10070973. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
8. Castelletti L., Yeo D., van Zandwijk N., Rasko J.E.J. Anti-Mesothelin CAR T cell therapy for malignant mesothelioma. Biomark. Res. 2021; 9 :11. doi: 10.1186/s40364-021-00264-1. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
9. Hassan R., Thomas A., Alewine C., Le D.T., Jaffee E.M., Pastan I. Mesothelin Immunotherapy for Cancer: Ready for Prime Time? J. Clin. Oncol. 2016; 34 :4171–4179. doi: 10.1200/JCO.2016.68.3672. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
10. Ye L., Lou Y., Lu L., Fan X. Mesothelin-targeted second generation CAR-T cells inhibit growth of mesothelin-expressing tumors in vivo. Exp. Ther. Med. 2019; 17 :739–747. doi: 10.3892/etm.2018.7015. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
11. Kaeding A.J., Barwe S.P., Gopalakrishnapillai A., Ries R.E., Alonzo T.A., Gerbing R.B., Correnti C., Loken M.R., Broderson L.E., Pardo L., et al. Mesothelin is a novel cell surface disease marker and potential therapeutic target in acute myeloid leukemia. Blood Adv. 2021; 5 :2350–2361. doi: 10.1182/bloodadvances.2021004424. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
12. Joseph S., Zhang X., Smith L.K., Alewine C. Furin is not required for processing of mesothelin precursor protein. Biochim. Biophys. Acta Mol. Cell. Res. 2021; 1868 :118967. doi: 10.1016/j.bbamcr.2021.118967. [ PubMed ] [ CrossRef ] [ Google Scholar ]
13. Yamaguchi N., Hattori K., Oh-eda M., Kojima T., Imai N., Ochi N. A novel cytokine exhibiting megakaryocyte potentiating activity from a human pancreatic tumor cell line HPC-Y5. J. Biol. Chem. 1994; 269 :805–808. doi: 10.1016/S0021-9258(17)42180-6. [ PubMed ] [ CrossRef ] [ Google Scholar ]
14. Weber D.G., Taeger D., Pesch B., Kraus T., Bruning T., Johnen G. Soluble mesothelin-related peptides (SMRP)—High stability of a potential tumor marker for mesothelioma. Cancer Biomark. 2007; 3 :287–292. doi: 10.3233/CBM-2007-3602. [ PubMed ] [ CrossRef ] [ Google Scholar ]
15. Grosso F., Mannucci M., Ugo F., Ferro P., Cassinari M., Vigani A., De Angelis A.M., Delfanti S., Lia M., Guaschino R., et al. Pilot Study to Evaluate Serum Soluble Mesothelin-Related Peptide (SMRP) as Marker for Clinical Monitoring of Pleural Mesothelioma (PM): Correlation with Modified RECIST Score. Diagnostics. 2021; 11 :2015. doi: 10.3390/diagnostics11112015. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
16. Liu X., Chan A., Tai C.H., Andresson T., Pastan I. Multiple proteases are involved in mesothelin shedding by cancer cells. Commun. Biol. 2020; 3 :728. doi: 10.1038/s42003-020-01464-5. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
17. Sathyanarayana B.K., Hahn Y., Patankar M.S., Pastan I., Lee B. Mesothelin, Stereocilin, and Otoancorin are predicted to have superhelical structures with ARM-type repeats. BMC Struct. Biol. 2009; 9 :1. doi: 10.1186/1472-6807-9-1. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
18. Ma J., Tang W.K., Esser L., Pastan I., Xia D. Recognition of mesothelin by the therapeutic antibody MORAb-009: Structural and mechanistic insights. J. Biol. Chem. 2012; 287 :33123–33131. doi: 10.1074/jbc.M112.381756. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
19. Dustin M.L., Selvaraj P., Mattaliano R.J., Springer T.A. Anchoring mechanisms for LFA-3 cell adhesion glycoprotein at membrane surface. Nature. 1987; 329 :846–848. doi: 10.1038/329846a0. [ PubMed ] [ CrossRef ] [ Google Scholar ]
20. Stefanova I., Horejsi V., Ansotegui I.J., Knapp W., Stockinger H. GPI-anchored cell-surface molecules complexed to protein tyrosine kinases. Science. 1991; 254 :1016–1019. doi: 10.1126/science.1719635. [ PubMed ] [ CrossRef ] [ Google Scholar ]
21. Bera T.K., Pastan I. Mesothelin is not required for normal mouse development or reproduction. Mol. Cell. Biol. 2000; 20 :2902–2906. doi: 10.1128/MCB.20.8.2902-2906.2000. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
22. Hilliard T.S., Kowalski B., Iwamoto K., Agadi E.A., Liu Y., Yang J., Asem M., Klymenko Y., Johnson J., Shi Z., et al. Host Mesothelin Expression Increases Ovarian Cancer Metastasis in the Peritoneal Microenvironment. Int. J. Mol. Sci. 2021; 22 :2443. doi: 10.3390/ijms222212443. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
23. Zhang J., Bera T.K., Liu W., Du X., Alewine C., Hassan R., Pastan I. Megakaryocytic potentiating factor and mature mesothelin stimulate the growth of a lung cancer cell line in the peritoneal cavity of mice. PLoS ONE. 2014; 9 :e104388. doi: 10.1371/journal.pone.0104388. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
24. Gubbels J.A., Belisle J., Onda M., Rancourt C., Migneault M., Ho M., Bera T.K., Connor J., Sathyanarayana B.K., Lee B., et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol. Cancer. 2006; 5 :50. doi: 10.1186/1476-4598-5-50. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
25. Rump A., Morikawa Y., Tanaka M., Minami S., Umesaki N., Takeuchi M., Miyajima A. Binding of ovarian cancer antigen CA125/MUC16 to mesothelin mediates cell adhesion. J. Biol. Chem. 2004; 279 :9190–9198. doi: 10.1074/jbc.M312372200. [ PubMed ] [ CrossRef ] [ Google Scholar ]
26. Bast R.C., Jr., Klug T.L., St John E., Jenison E., Niloff J.M., Lazarus H., Berkowitz R.S., Leavitt T., Griffiths C.T., Parker L., et al. A radioimmunoassay using a monoclonal antibody to monitor the course of epithelial ovarian cancer. N. Engl. J. Med. 1983; 309 :883–887. doi: 10.1056/NEJM198310133091503. [ PubMed ] [ CrossRef ] [ Google Scholar ]
27. Kaneko O., Gong L., Zhang J., Hansen J.K., Hassan R., Lee B., Ho M. A binding domain on mesothelin for CA125/MUC16. J. Biol. Chem. 2009; 284 :3739–3749. doi: 10.1074/jbc.M806776200. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
28. Huo Q., Xu C., Shao Y., Yu Q., Huang L., Liu Y., Bao H. Free CA125 promotes ovarian cancer cell migration and tumor metastasis by binding Mesothelin to reduce DKK1 expression and activate the SGK3/FOXO3 pathway. Int. J. Biol. Sci. 2021; 17 :574–588. doi: 10.7150/ijbs.52097. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
29. Chang M.C., Chen C.A., Chen P.J., Chiang Y.C., Chen Y.L., Mao T.L., Lin H.W., Lin Chiang W.H., Cheng W.F. Mesothelin enhances invasion of ovarian cancer by inducing MMP-7 through MAPK/ERK and JNK pathways. Biochem. J. 2012; 442 :293–302. doi: 10.1042/BJ20110282. [ PubMed ] [ CrossRef ] [ Google Scholar ]
30. Egeblad M., Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer. 2002; 2 :161–174. doi: 10.1038/nrc745. [ PubMed ] [ CrossRef ] [ Google Scholar ]
31. Chen S.H., Hung W.C., Wang P., Paul C., Konstantopoulos K. Mesothelin binding to CA125/MUC16 promotes pancreatic cancer cell motility and invasion via MMP-7 activation. Sci. Rep. 2013; 3 :1870. doi: 10.1038/srep01870. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
32. Dainty L.A., Risinger J.I., Morrison C., Chandramouli G.V., Bidus M.A., Zahn C., Rose G.S., Fowler J., Berchuck A., Maxwell G.L. Overexpression of folate binding protein and mesothelin are associated with uterine serous carcinoma. Gynecol. Oncol. 2007; 105 :563–570. doi: 10.1016/j.ygyno.2006.10.063. [ PubMed ] [ CrossRef ] [ Google Scholar ]
33. Galloway M.L., Murray D., Moffat D.F. The use of the monoclonal antibody mesothelin in the diagnosis of malignant mesothelioma in pleural biopsies. Histopathology. 2006; 48 :767–769. doi: 10.1111/j.1365-2559.2005.02279.x. [ PubMed ] [ CrossRef ] [ Google Scholar ]
34. Ho M., Bera T.K., Willingham M.C., Onda M., Hassan R., FitzGerald D., Pastan I. Mesothelin expression in human lung cancer. Clin. Cancer Res. 2007; 13 :1571–1575. doi: 10.1158/1078-0432.CCR-06-2161. [ PubMed ] [ CrossRef ] [ Google Scholar ]
35. Ordonez N.G. Application of mesothelin immunostaining in tumor diagnosis. Am. J. Surg. Pathol. 2003; 27 :1418–1428. doi: 10.1097/00000478-200311000-00003. [ PubMed ] [ CrossRef ] [ Google Scholar ]
36. Yu L., Feng M., Kim H., Phung Y., Kleiner D.E., Gores G.J., Qian M., Wang X.W., Ho M. Mesothelin as a potential therapeutic target in human cholangiocarcinoma. J. Cancer. 2010; 1 :141–149. doi: 10.7150/jca.1.141. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
37. Nagata K., Shinto E., Shiraishi T., Yamadera M., Kajiwara Y., Mochizuki S., Okamoto K., Einama T., Kishi Y., Ueno H. Mesothelin Expression is Correlated with Chemoresistance in Stage IV Colorectal Cancer. Ann. Surg. Oncol. 2021; 28 :8579–8586. doi: 10.1245/s10434-021-10507-y. [ PubMed ] [ CrossRef ] [ Google Scholar ]
38. Bharadwaj U., Li M., Chen C., Yao Q. Mesothelin-induced pancreatic cancer cell proliferation involves alteration of cyclin E via activation of signal transducer and activator of transcription protein 3. Mol. Cancer Res. 2008; 6 :1755–1765. doi: 10.1158/1541-7786.MCR-08-0095. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
39. Bharadwaj U., Marin-Muller C., Li M., Chen C., Yao Q. Mesothelin overexpression promotes autocrine IL-6/sIL-6R trans-signaling to stimulate pancreatic cancer cell proliferation. Carcinogenesis. 2011; 32 :1013–1024. doi: 10.1093/carcin/bgr075. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
40. Uehara N., Matsuoka Y., Tsubura A. Mesothelin promotes anchorage-independent growth and prevents anoikis via extracellular signal-regulated kinase signaling pathway in human breast cancer cells. Mol. Cancer Res. 2008; 6 :186–193. doi: 10.1158/1541-7786.MCR-07-0254. [ PubMed ] [ CrossRef ] [ Google Scholar ]
41. Weidemann S., Gagelmann P., Gorbokon N., Lennartz M., Menz A., Luebke A.M., Kluth M., Hube-Magg C., Blessin N.C., Fraune C., et al. Mesothelin Expression in Human Tumors: A Tissue Microarray Study on 12,679 Tumors. Biomedicines. 2021; 9 :397. doi: 10.3390/biomedicines9040397. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
42. Weidemann S., Perez D., Izbicki J.R., Neipp M., Mofid H., Daniels T., Nahrstedt U., Jacobsen F., Bernreuther C., Simon R., et al. Mesothelin is Commonly Expressed in Pancreatic Adenocarcinoma but Unrelated to Cancer Aggressiveness. Cancer Investig. 2021; 39 :711–720. doi: 10.1080/07357907.2021.1943747. [ PubMed ] [ CrossRef ] [ Google Scholar ]
43. Das V., Bhattacharya S., Chikkaputtaiah C., Hazra S., Pal M. The basics of epithelial-mesenchymal transition (EMT): A study from a structure, dynamics, and functional perspective. J. Cell. Physiol. 2019; 234 :14535–14555. doi: 10.1002/jcp.28160. [ PubMed ] [ CrossRef ] [ Google Scholar ]
44. He X., Wang L., Riedel H., Wang K., Yang Y., Dinu C.Z., Rojanasakul Y. Mesothelin promotes epithelial-to-mesenchymal transition and tumorigenicity of human lung cancer and mesothelioma cells. Mol. Cancer. 2017; 16 :63. doi: 10.1186/s12943-017-0633-8. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
45. Wang K., Bodempudi V., Liu Z., Borrego-Diaz E., Yamoutpoor F., Meyer A., Woo R.A., Pan W., Dudek A.Z., Olyaee M.S., et al. Inhibition of mesothelin as a novel strategy for targeting cancer cells. PLoS ONE. 2012; 7 :e33214. doi: 10.1371/journal.pone.0033214. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
46. Lurie E., Liu D., LaPlante E.L., Thistlethwaite L.R., Yao Q., Milosavljevic A. Histoepigenetic analysis of the mesothelin network within pancreatic ductal adenocarcinoma cells reveals regulation of retinoic acid receptor gamma and AKT by mesothelin. Oncogenesis. 2020; 9 :62. doi: 10.1038/s41389-020-00245-3. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
47. Tang Z., Qian M., Ho M. The role of mesothelin in tumor progression and targeted therapy. Anticancer Agents Med. Chem. 2013; 13 :276–280. doi: 10.2174/1871520611313020014. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
48. Yang X., Huang M., Zhang Q., Chen J., Li J., Han Q., Zhang L., Li J., Liu S., Ma Y., et al. Metformin Antagonizes Ovarian Cancer Cells Malignancy Through MSLN Mediated IL-6/STAT3 Signaling. Cell Transplant. 2021; 30 :9636897211027819. doi: 10.1177/09636897211027819. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
49. Paulick M.G., Bertozzi C.R. The glycosylphosphatidylinositol anchor: A complex membrane-anchoring structure for proteins. Biochemistry. 2008; 47 :6991–7000. doi: 10.1021/bi8006324. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
50. Dangaj D., Abbott K.L., Mookerjee A., Zhao A., Kirby P.S., Sandaltzopoulos R., Powell D.J., Jr., Lamaziere A., Siegel D.L., Wolf C., et al. Mannose receptor (MR) engagement by mesothelin GPI anchor polarizes tumor-associated macrophages and is blocked by anti-MR human recombinant antibody. PLoS ONE. 2011; 6 :e28386. doi: 10.1371/journal.pone.0028386. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
51. Hucl T., Brody J.R., Gallmeier E., Iacobuzio-Donahue C.A., Farrance I.K., Kern S.E. High cancer-specific expression of mesothelin (MSLN) is attributable to an upstream enhancer containing a transcription enhancer factor dependent MCAT motif. Cancer Res. 2007; 67 :9055–9065. doi: 10.1158/0008-5472.CAN-07-0474. [ PubMed ] [ CrossRef ] [ Google Scholar ]
52. Muminova Z.E., Strong T.V., Shaw D.R. Characterization of human mesothelin transcripts in ovarian and pancreatic cancer. BMC Cancer. 2004; 4 :19. doi: 10.1186/1471-2407-4-19. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
53. Hollevoet K., Mason-Osann E., Muller F., Pastan I. Methylation-associated partial down-regulation of mesothelin causes resistance to anti-mesothelin immunotoxins in a pancreatic cancer cell line. PLoS ONE. 2015; 10 :e0122462. doi: 10.1371/journal.pone.0122462. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
54. Sato N., Maitra A., Fukushima N., van Heek N.T., Matsubayashi H., Iacobuzio-Donahue C.A., Rosty C., Goggins M. Frequent hypomethylation of multiple genes overexpressed in pancreatic ductal adenocarcinoma. Cancer Res. 2003; 63 :4158–4166. [ PubMed ] [ Google Scholar ]
55. Tan K., Kajino K., Momose S., Masaoka A., Sasahara K., Shiomi K., Izumi H., Abe M., Ohtsuji N., Wang T., et al. Mesothelin (MSLN) promoter is hypomethylated in malignant mesothelioma, but its expression is not associated with methylation status of the promoter. Hum. Pathol. 2010; 41 :1330–1338. doi: 10.1016/j.humpath.2010.03.002. [ PubMed ] [ CrossRef ] [ Google Scholar ]
56. De Santi C., Vencken S., Blake J., Haase B., Benes V., Gemignani F., Landi S., Greene C.M. Identification of MiR-21-5p as a Functional Regulator of Mesothelin Expression Using MicroRNA Capture Affinity Coupled with Next Generation Sequencing. PLoS ONE. 2017; 12 :e0170999. doi: 10.1371/journal.pone.0170999. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
57. Marin-Muller C., Li D., Bharadwaj U., Li M., Chen C., Hodges S.E., Fisher W.E., Mo Q., Hung M.C., Yao Q. A tumorigenic factor interactome connected through tumor suppressor microRNA-198 in human pancreatic cancer. Clin. Cancer Res. 2013; 19 :5901–5913. doi: 10.1158/1078-0432.CCR-12-3776. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
58. Prieve M.G., Moon R.T. Stromelysin-1 and mesothelin are differentially regulated by Wnt-5a and Wnt-1 in C57mg mouse mammary epithelial cells. BMC Dev. Biol. 2003; 3 :2. doi: 10.1186/1471-213X-3-2. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
59. Pastan I., Hassan R. Discovery of mesothelin and exploiting it as a target for immunotherapy. Cancer Res. 2014; 74 :2907–2912. doi: 10.1158/0008-5472.CAN-14-0337. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
60. Kelly R.J., Sharon E., Pastan I., Hassan R. Mesothelin-targeted agents in clinical trials and in preclinical development. Mol. Cancer Ther. 2012; 11 :517–525. doi: 10.1158/1535-7163.MCT-11-0454. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
61. Le Q., Castro S., Tang T., Loeb A.M., Hylkema T., McKay C.N., Perkins L., Srivastava S., Call L., Smith J., et al. Therapeutic Targeting of Mesothelin with Chimeric Antigen Receptor T Cells in Acute Myeloid Leukemia. Clin. Cancer Res. 2021; 27 :5718–5730. doi: 10.1158/1078-0432.CCR-21-1546. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
62. Tang T.T., Call L.F., Castro S., Nourigat-Mckay C., Perkins L., Pardo L., Leonti A.R., Meshinchi S., Le Q. Therapeutic Targeting of Mesothelin with Chimeric Antigen Receptor Natural Killer Cell Therapy in Acute Myeloid Leukemia. Blood. 2021; 138 :1712. doi: 10.1182/blood-2021-153156. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
63. Gopalakrishnapillai A., Correnti C.E., Pilat K., Lin I., Chan M.K., Bandaranayake A.D., Mehlin C., Kisielewski A., Hamill D., Kaeding A.J., et al. Immunotherapeutic Targeting of Mesothelin Positive Pediatric AML Using Bispecific T Cell Engaging Antibodies. Cancers. 2021; 13 :5964. doi: 10.3390/cancers13235964. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
64. Wang Z., Li N., Feng K., Chen M., Zhang Y., Liu Y., Yang Q., Nie J., Tang N., Zhang X., et al. Phase I study of CAR-T cells with PD-1 and TCR disruption in mesothelin-positive solid tumors. Cell. Mol. Immunol. 2021; 18 :2188–2198. doi: 10.1038/s41423-021-00749-x. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
65. Haas A.R., Tanyi J.L., O’Hara M.H., Gladney W.L., Lacey S.F., Torigian D.A., Soulen M.C., Tian L., McGarvey M., Nelson A.M., et al. Phase I Study of Lentiviral-Transduced Chimeric Antigen Receptor-Modified T Cells Recognizing Mesothelin in Advanced Solid Cancers. Mol. Ther. 2019; 27 :1919–1929. doi: 10.1016/j.ymthe.2019.07.015. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
66. Pegna G.J., Ahmad M.I., Steinberg S.M., Peer C.J., Figg W.D., FitzGerald D.J., Hassan R., Pastan I., Alewine C. Phase I study of mesothelin-targeted immunotoxin LMB-100 in combination with tofacitinib in persons with pancreatobiliary cancer or other mesothelin expressing solid tumors. J. Clin. Oncol. 2021; 39 :TPS452. doi: 10.1200/JCO.2021.39.3_suppl.TPS452. [ CrossRef ] [ Google Scholar ]
67. Hagemann U.B., Ellingsen C., Schuhmacher J., Kristian A., Mobergslien A., Cruciani V., Wickstroem K., Schatz C.A., Kneip C., Golfier S., et al. Mesothelin-Targeted Thorium-227 Conjugate (MSLN-TTC): Preclinical Evaluation of a New Targeted Alpha Therapy for Mesothelin-Positive Cancers. Clin. Cancer Res. 2019; 25 :4723–4734. doi: 10.1158/1078-0432.CCR-18-3476. [ PubMed ] [ CrossRef ] [ Google Scholar ]
68. Tsujikawa T., Crocenzi T., Durham J.N., Sugar E.A., Wu A.A., Onners B., Nauroth J.M., Anders R.A., Fertig E.J., Laheru D.A., et al. Evaluation of Cyclophosphamide/GVAX Pancreas Followed by Listeria-Mesothelin (CRS-207) with or without Nivolumab in Patients with Pancreatic Cancer. Clin. Cancer Res. 2020; 26 :3578–3588. doi: 10.1158/1078-0432.CCR-19-3978. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
69. Han Y., Liu D., Li L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020; 10 :727–742. [ PMC free article ] [ PubMed ] [ Google Scholar ]
70. Chowdhury P.S., Viner J.L., Beers R., Pastan I. Isolation of a high-affinity stable single-chain Fv specific for mesothelin from DNA-immunized mice by phage display and construction of a recombinant immunotoxin with anti-tumor activity. Proc. Natl. Acad. Sci. USA. 1998; 95 :669–674. doi: 10.1073/pnas.95.2.669. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
71. Hassan R., Miller A.C., Sharon E., Thomas A., Reynolds J.C., Ling A., Kreitman R.J., Miettinen M.M., Steinberg S.M., Fowler D.H., et al. Major Cancer Regressions in Mesothelioma after Treatment with an Anti-Mesothelin Immunotoxin and Immune Suppression. Sci. Transl. Med. 2013; 5 :208ra147. doi: 10.1126/scitranslmed.3006941. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
72. Hollevoet K., Mason-Osann E., Liu X.F., Imhof-Jung S., Niederfellner G., Pastan I. In vitro and in vivo activity of the low-immunogenic antimesothelin immunotoxin RG7787 in pancreatic cancer. Mol. Cancer Ther. 2014; 13 :2040–2049. doi: 10.1158/1535-7163.MCT-14-0089-T. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
73. Hassan R., Alewine C., Mian I., Spreafico A., Siu L.L., Gomez-Roca C., Delord J.P., Italiano A., Lassen U., Soria J.C., et al. Phase 1 study of the immunotoxin LMB-100 in patients with mesothelioma and other solid tumors expressing mesothelin. Cancer. 2020; 126 :4936–4947. doi: 10.1002/cncr.33145. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
74. Alewine C., Ahmad M., Peer C.J., Hu Z.I., Lee M.J., Yuno A., Kindrick J.D., Thomas A., Steinberg S.M., Trepel J.B., et al. Phase I/II Study of the Mesothelin-targeted Immunotoxin LMB-100 with Nab-Paclitaxel for Patients with Advanced Pancreatic Adenocarcinoma. Clin. Cancer Res. 2020; 26 :828–836. doi: 10.1158/1078-0432.CCR-19-2586. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
75. Jiang Q., Ghafoor A., Mian I., Rathkey D., Thomas A., Alewine C., Sengupta M., Ahlman M.A., Zhang J., Morrow B., et al. Enhanced efficacy of mesothelin-targeted immunotoxin LMB-100 and anti-PD-1 antibody in patients with mesothelioma and mouse tumor models. Sci. Transl. Med. 2020; 12 :eaaz7252. doi: 10.1126/scitranslmed.aaz7252. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
76. Hassan R., Ebel W., Routhier E.L., Patel R., Kline J.B., Zhang J., Chao Q., Jacob S., Turchin H., Gibbs L., et al. Preclinical evaluation of MORAb-009, a chimeric antibody targeting tumor-associated mesothelin. Cancer Immun. Arch. 2007; 7 :20. [ PMC free article ] [ PubMed ] [ Google Scholar ]
77. Hassan R., Kindler H.L., Jahan T., Bazhenova L., Reck M., Thomas A., Pastan I., Parno J., O’Shannessy D.J., Fatato P., et al. Phase II clinical trial of amatuximab, a chimeric antimesothelin antibody with pemetrexed and cisplatin in advanced unresectable pleural mesothelioma. Clin. Cancer Res. 2014; 20 :5927–5936. doi: 10.1158/1078-0432.CCR-14-0804. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
78. Hassan R., Cohen S.J., Phillips M., Pastan I., Sharon E., Kelly R.J., Schweizer C., Weil S., Laheru D. Phase I clinical trial of the chimeric anti-mesothelin monoclonal antibody MORAb-009 in patients with mesothelin-expressing cancers. Clin. Cancer Res. 2010; 16 :6132–6138. doi: 10.1158/1078-0432.CCR-10-2275. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
79. Matsuzawa F., Kamachi H., Mizukami T., Einama T., Kawamata F., Fujii Y., Fukai M., Kobayashi N., Hatanaka Y., Taketomi A. Mesothelin blockage by Amatuximab suppresses cell invasiveness, enhances gemcitabine sensitivity and regulates cancer cell stemness in mesothelin-positive pancreatic cancer cells. BMC Cancer. 2021; 21 :200. doi: 10.1186/s12885-020-07722-3. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
80. Fujii Y., Kamachi H., Matsuzawa F., Mizukami T., Kobayashi N., Fukai M., Taketomi A. Early administration of amatuximab, a chimeric high-affinity anti-mesothelin monoclonal antibody, suppresses liver metastasis of mesothelin-expressing pancreatic cancer cells and enhances gemcitabine sensitivity in a xenograft mouse model. Investig. New Drugs. 2021; 39 :1256–1266. doi: 10.1007/s10637-021-01118-1. [ PubMed ] [ CrossRef ] [ Google Scholar ]
81. Jaffee E.M., Hruban R.H., Biedrzycki B., Laheru D., Schepers K., Sauter P.R., Goemann M., Coleman J., Grochow L., Donehower R.C., et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: A phase I trial of safety and immune activation. J. Clin. Oncol. 2001; 19 :145–156. doi: 10.1200/JCO.2001.19.1.145. [ PubMed ] [ CrossRef ] [ Google Scholar ]
82. Le D.T., Brockstedt D.G., Nir-Paz R., Hampl J., Mathur S., Nemunaitis J., Sterman D.H., Hassan R., Lutz E., Moyer B., et al. A live-attenuated Listeria vaccine (ANZ-100) and a live-attenuated Listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: Phase I studies of safety and immune induction. Clin. Cancer Res. 2012; 18 :858–868. doi: 10.1158/1078-0432.CCR-11-2121. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
83. Thomas A.M., Santarsiero L.M., Lutz E.R., Armstrong T.D., Chen Y.C., Huang L.Q., Laheru D.A., Goggins M., Hruban R.H., Jaffee E.M. Mesothelin-specific CD8(+) T cell responses provide evidence of in vivo cross-priming by antigen-presenting cells in vaccinated pancreatic cancer patients. J. Exp. Med. 2004; 200 :297–306. doi: 10.1084/jem.20031435. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
84. Le D.T., Wang-Gillam A., Picozzi V., Greten T.F., Crocenzi T., Springett G., Morse M., Zeh H., Cohen D., Fine R.L., et al. Safety and survival with GVAX pancreas prime and Listeria Monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J. Clin. Oncol. 2015; 33 :1325–1333. doi: 10.1200/JCO.2014.57.4244. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
85. Le D.T., Picozzi V.J., Ko A.H., Wainberg Z.A., Kindler H., Wang-Gillam A., Oberstein P., Morse M.A., Zeh H.J., 3rd, Weekes C., et al. Results from a Phase IIb, Randomized, Multicenter Study of GVAX Pancreas and CRS-207 Compared with Chemotherapy in Adults with Previously Treated Metastatic Pancreatic Adenocarcinoma (ECLIPSE Study) Clin. Cancer Res. 2019; 25 :5493–5502. doi: 10.1158/1078-0432.CCR-18-2992. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
86. Hassan R., Alley E., Kindler H., Antonia S., Jahan T., Honarmand S., Nair N., Whiting C.C., Enstrom A., Lemmens E., et al. Clinical Response of Live-Attenuated, Listeria monocytogenes Expressing Mesothelin (CRS-207) with Chemotherapy in Patients with Malignant Pleural Mesothelioma. Clin. Cancer Res. 2019; 25 :5787–5798. doi: 10.1158/1078-0432.CCR-19-0070. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
87. Golfier S., Kopitz C., Kahnert A., Heisler I., Schatz C.A., Stelte-Ludwig B., Mayer-Bartschmid A., Unterschemmann K., Bruder S., Linden L., et al. Anetumab ravtansine: A novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol. Cancer Ther. 2014; 13 :1537–1548. doi: 10.1158/1535-7163.MCT-13-0926. [ PubMed ] [ CrossRef ] [ Google Scholar ]
88. Hassan R., Blumenschein G.R., Jr., Moore K.N., Santin A.D., Kindler H.L., Nemunaitis J.J., Seward S.M., Thomas A., Kim S.K., Rajagopalan P., et al. First-in-Human, Multicenter, Phase I Dose-Escalation and Expansion Study of Anti-Mesothelin Antibody-Drug Conjugate Anetumab Ravtansine in Advanced or Metastatic Solid Tumors. J. Clin. Oncol. 2020; 38 :1824–1835. doi: 10.1200/JCO.19.02085. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
89. Quanz M., Hagemann U.B., Zitzmann-Kolbe S., Stelte-Ludwig B., Golfier S., Elbi C., Mumberg D., Ziegelbauer K., Schatz C.A. Anetumab ravtansine inhibits tumor growth and shows additive effect in combination with targeted agents and chemotherapy in mesothelin-expressing human ovarian cancer models. Oncotarget. 2018; 9 :34103–34121. doi: 10.18632/oncotarget.26135. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
90. Weekes C.D., Lamberts L.E., Borad M.J., Voortman J., McWilliams R.R., Diamond J.R., de Vries E.G., Verheul H.M., Lieu C.H., Kim G.P., et al. Phase I Study of DMOT4039A, an Antibody-Drug Conjugate Targeting Mesothelin, in Patients with Unresectable Pancreatic or Platinum-Resistant Ovarian Cancer. Mol. Cancer Ther. 2016; 15 :439–447. doi: 10.1158/1535-7163.MCT-15-0693. [ PubMed ] [ CrossRef ] [ Google Scholar ]
91. Rottey S., Clarke J., Aung K., Machiels J.P., Markman B., Heinhuis K.M., Millward M., Lolkema M., Patel S.P., de Souza P., et al. Phase 1/2a trial of BMS-986148, an anti-mesothelin antibody-drug conjugate, alone or in combination with nivolumab in patients with advanced solid tumors. Clin. Cancer Res. 2021; 28 :95–105. doi: 10.1158/1078-0432.CCR-21-1181. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
92. Sadelain M., Brentjens R., Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013; 3 :388–398. doi: 10.1158/2159-8290.CD-12-0548. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
93. O’Hara M., Stashwick C., Haas A.R., Tanyi J.L. Mesothelin as a target for chimeric antigen receptor-modified T cells as anticancer therapy. Immunotherapy. 2016; 8 :449–460. doi: 10.2217/imt.16.4. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
94. Morello A., Sadelain M., Adusumilli P.S. Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Discov. 2016; 6 :133–146. doi: 10.1158/2159-8290.CD-15-0583. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
95. Lanitis E., Poussin M., Hagemann I.S., Coukos G., Sandaltzopoulos R., Scholler N., Powell D.J., Jr. Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptor. Mol. Ther. 2012; 20 :633–643. doi: 10.1038/mt.2011.256. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
96. Klampatsa A., Dimou V., Albelda S.M. Mesothelin-targeted CAR-T cell therapy for solid tumors. Expert Opin. Biol. Ther. 2021; 21 :473–486. doi: 10.1080/14712598.2021.1843628. [ PubMed ] [ CrossRef ] [ Google Scholar ]
97. Ding J., Horton M., Shah S., Zieba A., Krishnamurphy J., Ashhurst T., Menk A.V., Baeuerle P.A., King N.J., Delogoffe G., et al. Preclinical evaluation of TC-210, a mesothelin-specific T cell receptor (TCR) fusion construct (TRuC™) T cells for the treatment of solid tumors. Cancer Res. 2019; 79 :2307. [ Google Scholar ]
98. Tong C., Zhang Y., Liu Y., Ji X., Zhang W., Guo Y., Han X., Ti D., Dai H., Wang C., et al. Optimized tandem CD19/CD20 CAR-engineered T cells in refractory/relapsed B-cell lymphoma. Blood. 2020; 136 :1632–1644. doi: 10.1182/blood.2020005278. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
99. Han X., Wang Y., Wei J., Han W. Multi-antigen-targeted chimeric antigen receptor T cells for cancer therapy. J. Hematol. Oncol. 2019; 12 :128. doi: 10.1186/s13045-019-0813-7. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
100. Liang Z., Dong J., Yang N., Li S.D., Yang Z.Y., Huang R., Li F.J., Wang W.T., Ren J.K., Lei J., et al. Tandem CAR-T cells targeting FOLR1 and MSLN enhance the antitumor effects in ovarian cancer. Int. J. Biol. Sci. 2021; 17 :4365–4376. doi: 10.7150/ijbs.63181. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
101. Liu G., Zhang Q., Li D., Zhang L., Gu Z., Liu J., Liu G., Yang M., Gu J., Cui X., et al. PD-1 silencing improves anti-tumor activities of human mesothelin-targeted CAR T cells. Hum. Immunol. 2021; 82 :130–138. doi: 10.1016/j.humimm.2020.12.002. [ PubMed ] [ CrossRef ] [ Google Scholar ]
102. Rezaei M., Tan J., Zeng C., Li Y., Ganjalikhani-Hakemi M. TIM-3 in Leukemia; Immune Response and Beyond. Front. Oncol. 2021; 11 :753677. doi: 10.3389/fonc.2021.753677. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
103. Liu J.F., Wu L., Yang L.L., Deng W.W., Mao L., Wu H., Zhang W.F., Sun Z.J. Blockade of TIM3 relieves immunosuppression through reducing regulatory T cells in head and neck cancer. J. Exp. Clin. Cancer Res. 2018; 37 :44. doi: 10.1186/s13046-018-0713-7. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
104. Jafarzadeh L., Masoumi E., Mirzaei H.R., Alishah K., Fallah-Mehrjardi K., Khakpoor-Koosheh M., Rostamian H., Noorbakhsh F., Hadjati J. Targeted knockdown of Tim3 by short hairpin RNAs improves the function of anti-mesothelin CAR T cells. Mol. Immunol. 2021; 139 :1–9. doi: 10.1016/j.molimm.2021.06.007. [ PubMed ] [ CrossRef ] [ Google Scholar ]
105. Liu G., Zhang Q., Liu G., Li D., Zhang L., Gu Z., Tian H., Zhang Y., Tian X. Disruption of adenosine 2A receptor improves the anti-tumor function of anti-mesothelin CAR T cells both in vitro and in vivo. Exp. Cell Res. 2021; 409 :112886. doi: 10.1016/j.yexcr.2021.112886. [ PubMed ] [ CrossRef ] [ Google Scholar ]
106. Wang Y., Wang J., Yang X., Yang J., Lu P., Zhao L., Li B., Pan H., Jiang Z., Shen X., et al. Chemokine Receptor CCR2b Enhanced Anti-tumor Function of Chimeric Antigen Receptor T Cells Targeting Mesothelin in a Non-small-cell Lung Carcinoma Model. Front. Immunol. 2021; 12 :628906. doi: 10.3389/fimmu.2021.628906. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
107. Cao B., Liu M., Wang L., Liang B., Feng Y., Chen X., Shi Y., Zhang J., Ye X., Tian Y., et al. Use of chimeric antigen receptor NK-92 cells to target mesothelin in ovarian cancer. Biochem. Biophys. Res. Commun. 2020; 524 :96–102. doi: 10.1016/j.bbrc.2020.01.053. [ PubMed ] [ CrossRef ] [ Google Scholar ]
108. Cao B., Liu M., Huang J., Zhou J., Li J., Lian H., Huang W., Guo Y., Yang S., Lin L., et al. Development of mesothelin-specific CAR NK-92 cells for the treatment of gastric cancer. Int. J. Biol. Sci. 2021; 17 :3850–3861. doi: 10.7150/ijbs.64630. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
109. Velasquez M.P., Bonifant C.L., Gottschalk S. Redirecting T cells to hematological malignancies with bispecific antibodies. Blood. 2018; 131 :30–38. doi: 10.1182/blood-2017-06-741058. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
110. Del Bano J., Flores-Flores R., Josselin E., Goubard A., Ganier L., Castellano R., Chames P., Baty D., Kerfelec B. A Bispecific Antibody-Based Approach for Targeting Mesothelin in Triple Negative Breast Cancer. Front. Immunol. 2019; 10 :1593. doi: 10.3389/fimmu.2019.01593. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
111. Jurcak N.R., Zarecki M., Lee F., Rozich N., Muth S., Jaffee E., Stienen S., Bailis J., Zheng L. Abstract 1561: Evaluation of mesothelin BiTE® antibody constructs in models of pancreatic ductal adenocarcinoma. Cancer Res. 2019; 79 :1561. doi: 10.1158/1538-7445.Am2019-1561. [ CrossRef ] [ Google Scholar ]
112. Suurs F.V., Lorenczewski G., Bailis J.M., Stienen S., Friedrich M., Lee F., van der Vegt B., de Vries E.G.E., de Groot D.A., Lub-de Hooge M.N. Mesothelin/CD3 half-life extended bispecific T-cell engager molecule shows specific tumor uptake and distributes to mesothelin and CD3 expressing tissues. J. Nucl. Med. 2021; 62 :1797–1804. doi: 10.2967/jnumed.120.259036. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
113. Yoon A., Lee S., Lee S., Lim S., Park Y.Y., Song E., Kim D.S., Kim K., Lim Y. A Novel T Cell-Engaging Bispecific Antibody for Treating Mesothelin-Positive Solid Tumors. Biomolecules. 2020; 10 :399. doi: 10.3390/biom10030399. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
114. Molloy M.E., Austin R.J., Lemon B.D., Aaron W.H., Ganti V., Jones A., Jones S.D., Strobel K.L., Patnaik P., Sexton K., et al. Preclinical Characterization of HPN536, a Trispecific, T-Cell-Activating Protein Construct for the Treatment of Mesothelin-Expressing Solid Tumors. Clin. Cancer Res. 2021; 27 :1452–1462. doi: 10.1158/1078-0432.CCR-20-3392. [ PubMed ] [ CrossRef ] [ Google Scholar ]
115. Targeted Alpha Therapy Working Group. Parker C., Lewington V., Shore N., Kratochwil C., Levy M., Linden O., Noordzij W., Park J., Saad F. Targeted Alpha Therapy, an Emerging Class of Cancer Agents: A Review. JAMA Oncol. 2018; 4 :1765–1772. doi: 10.1001/jamaoncol.2018.4044. [ PubMed ] [ CrossRef ] [ Google Scholar ]
116. Wickstroem K., Hagemann U.B., Cruciani V., Wengner A.M., Kristian A., Ellingsen C., Siemeister G., Bjerke R.M., Karlsson J., Ryan O.B., et al. Synergistic Effect of a Mesothelin-Targeted (227)Th Conjugate in Combination with DNA Damage Response Inhibitors in Ovarian Cancer Xenograft Models. J. Nucl. Med. 2019; 60 :1293–1300. doi: 10.2967/jnumed.118.223701. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
117. Steinbach D., Schramm A., Eggert A., Onda M., Dawczynski K., Rump A., Pastan I., Wittig S., Pfaffendorf N., Voigt A., et al. Identification of a set of seven genes for the monitoring of minimal residual disease in pediatric acute myeloid leukemia. Clin. Cancer Res. 2006; 12 :2434–2441. doi: 10.1158/1078-0432.CCR-05-2552. [ PubMed ] [ CrossRef ] [ Google Scholar ]
118. Steinbach D., Onda M., Voigt A., Dawczynski K., Wittig S., Hassan R., Gruhn B., Pastan I. Mesothelin, a possible target for immunotherapy, is expressed in primary AML cells. Eur. J. Haematol. 2007; 79 :281–286. doi: 10.1111/j.1600-0609.2007.00928.x. [ PubMed ] [ CrossRef ] [ Google Scholar ]
119. Gopalakrishnapillai A., Kaeding A.J., Schatz C.A., Sommer A., Meshinchi S., Kolb E.A., Barwe S.P. In Vivo Evaluation of Mesothelin As a Therapeutic Target in Pediatric Acute Myeloid Leukemia. Blood. 2019; 134 :1370. doi: 10.1182/blood-2019-123794. [ CrossRef ] [ Google Scholar ]
120. Chen J., Glasser C.L. New and Emerging Targeted Therapies for Pediatric Acute Myeloid Leukemia (AML) Children. 2020; 7 :12. doi: 10.3390/children7020012. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
121. Tarlock K., Kaeding A.J., Alonzo T.A. Discovery and Validation of Cell-Surface Protein Mesothelin (MSLN) As a Novel Therapeutic Target in AML: Results from the COG/NCI Target AML Initiative. Blood. 2016; 128 :2873. doi: 10.1182/blood.V128.22.2873.2873. [ CrossRef ] [ Google Scholar ]
122. Tarlock K., Alonzo T.A., Gerbing R.B., Le Q., Tang T.T., Hamill D., Gopalakrishnapillai A., Barwe S.P., Ries R.E., Aplenc R., et al. Mesothelin Expression Is Associated with Extramedullary Disease and Promotes In Vivo Leukemic Growth in Acute Myeloid Leukemia. Blood. 2020; 136 :38–39. doi: 10.1182/blood-2020-134821. [ CrossRef ] [ Google Scholar ]

Articles from Cancers are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)