Combination Therapy of Immunotoxin and Checkpoint Inhibitor

Bigner; Darell ;   et al.

Patent Application Summary

U.S. patent application number 17/376319 was filed with the patent office on 2021-11-04 for combination therapy of immunotoxin and checkpoint inhibitor. The applicant listed for this patent is Duke University, THE GOVERNMENT OF THE UNITED STATES AS REPRESENTED BY THE SECRETARY OF HEALTH AND HUMAN SERVICES, THE GOVERNMENT OF THE UNITED STATES AS REPRESENTED BY THE SECRETARY OF HEALTH AND HUMAN SERVICES. Invention is credited to Xuhui Bao, Darell Bigner, Vidyalakshmi Chandramohan, Matthias Gromeier, Smita Nair, Ira H. Pastan.

Application Number20210338811 17/376319
Document ID /
Family ID1000005720461
Filed Date2021-11-04

United States Patent Application 20210338811
Kind Code A1
Bigner; Darell ;   et al. November 4, 2021

Combination Therapy of Immunotoxin and Checkpoint Inhibitor

Abstract

Regional, tumor-targeted, cytotoxic therapy, such as D2C7-immunotoxin (D2C7-IT), not only specifically target and destroy tumor cells, but in the process initiate immune events that promote an in situ vaccine effect. The antitumor effects are amplified by immune checkpoint blockade which engenders a long-term systemic immune response that effectively eliminates all tumor cells.


Inventors: Bigner; Darell; (Mebane, NC) ; Chandramohan; Vidyalakshmi; (Durham, NC) ; Nair; Smita; (Durham, NC) ; Gromeier; Matthias; (Durham, NC) ; Bao; Xuhui; (Durham, NC) ; Pastan; Ira H.; (Potomac, MD)
Applicant:
Name City State Country Type

Duke University
THE GOVERNMENT OF THE UNITED STATES AS REPRESENTED BY THE SECRETARY OF HEALTH AND HUMAN SERVICES

Durham
Rockville

NC
MD

US
US
Family ID: 1000005720461
Appl. No.: 17/376319
Filed: July 15, 2021

Related U.S. Patent Documents

Application Number Filing Date Patent Number
15773418 May 3, 2018 11065332
PCT/US16/60469 Nov 4, 2016
17376319
62250749 Nov 4, 2015

Current U.S. Class: 1/1
Current CPC Class: A61K 2039/507 20130101; C07K 2317/76 20130101; A61P 35/00 20180101; C07K 16/2818 20130101; A61K 39/395 20130101; A61K 47/6849 20170801; A61K 39/3955 20130101; A61K 47/6851 20170801; C07K 16/2863 20130101; C07K 2317/73 20130101; C07K 2317/624 20130101; C07K 2319/55 20130101; A61K 47/6829 20170801; C07K 2317/622 20130101; C07K 16/30 20130101; A61K 2039/505 20130101; A61K 38/164 20130101
International Class: A61K 39/395 20060101 A61K039/395; C07K 16/28 20060101 C07K016/28; C07K 16/30 20060101 C07K016/30; A61K 47/68 20060101 A61K047/68; A61P 35/00 20060101 A61P035/00; A61K 38/16 20060101 A61K038/16

Goverment Interests



[0001] This invention was made with government support under CA197264 and CA-154291 awarded by the National Institutes of Health. The government has certain rights in the invention.
Claims



1. A method of treating a tumor in a patient, comprising: administering to the patient an immunotoxin comprising a single chain variable region antibody fused to a PE38 truncated Pseudomonas exotoxin, wherein the single chain variable region antibody has CDR1, CDR2, and CDR3 regions as shown in SEQ ID NO: 6-11; and administering an immune checkpoint inhibitor to the patient.

2. The method of claim 1 wherein the tumor is a malignant glioma.

3. The method of claim 1 wherein the tumor is breast cancer.

4. The method of claim 1 wherein the tumor is head and neck squamous cell carcinoma.

5. The method of claim 1 wherein the tumor is lung cancer.

6. The method of claim 1 wherein the immunotoxin is administered directly to the tumor.

7. The method of claim 1 wherein the immune checkpoint is selected from the group consisting of PD-1, PD-L1, CTLA-4, LAG-3, TIM-3, and CSF-1R.

8. The method of claim 1 wherein the checkpoint inhibitor is an anti-PD-1 antibody.

9. The method of claim 1 wherein the checkpoint inhibitor is an anti-PD-L1 antibody.

10. The method of claim 1 wherein the checkpoint inhibitor is an anti-CTLA4 antibody.

11. The method of claim 1 wherein the checkpoint inhibitor is an anti-LAG-3 antibody.

12. The method of claim 1 wherein the checkpoint inhibitor is an anti-TIM-3 antibody.

13. The method of claim 1 wherein the checkpoint inhibitor is an anti-CSF-1R antibody.

14. The method of claim 1 wherein the checkpoint inhibitor is a small molecule inhibitor of IDO.

15. The method of claim 1 wherein the checkpoint inhibitor is a small molecule inhibitor of CSF-1R.

16. The method of claim 1 wherein the immune checkpoint inhibitor is administered within 30 days of administering the immunotoxin.

17. The method of claim 1 wherein the immune checkpoint inhibitor is administered within 7 days of administering the immunotoxin.

18. The method of claim 1 wherein the PE38 truncated Pseudomonas exotoxin is fused to a KDEL peptide.

19. A kit for treating a tumor, comprising: an immunotoxin comprising a single chain variable region antibody fused to a PE38 truncated Pseudomonas exotoxin, wherein the single chain variable region antibody has CDR1, CDR2, and CDR3 regions as shown in SEQ ID NO: 6-11; and an immune checkpoint inhibitor.

20. The kit of claim 19 wherein the checkpoint inhibitor is an anti-PD-1 antibody.

21. The kit of claim 19 wherein the checkpoint inhibitor is an anti-PDL-1 antibody.

22. The kit of claim 19 wherein the checkpoint inhibitor is an anti-CTLA4 antibody.

23. The kit of claim 19 wherein the checkpoint inhibitor is an anti-LAG-3 antibody.

24. The kit of claim 19 wherein the checkpoint inhibitor is an anti-TIM-3 antibody.

25. The kit of claim 19 wherein the checkpoint inhibitor is an anti-CSF-R1 antibody.

26. The kit of claim 19 wherein the checkpoint inhibitor is a small molecule inhibitor of IDO.

27. The kit of claim 19 wherein the checkpoint inhibitor is a small molecule inhibitor of CSF-R1.

28. The kit of claim 19 wherein the PE38 truncated Pseudomonas exotoxin is fused to a KDEL peptide.
Description



TECHNICAL FIELD OF THE INVENTION

[0002] This invention is related to the area of immunotherapy. In particular, it relates to combination regimens for treating tumors and kits and medicaments for accomplishing them.

BACKGROUND OF THE INVENTION

[0003] Glioblastoma is the most dismal malignant brain tumor among all primary brain and central nervous system tumors. The median survival time for glioblastoma patients with the current standard treatment or even newly developed agents is less than 15 months. Thus, there is an urgent need to develop advanced and efficient therapeutic approaches to improve the poor survival outlook of glioblastoma patients as well as other tumors expressing EGFR receptors.

SUMMARY OF THE INVENTION

[0004] According to one aspect of the invention a method is provided for treating a tumor in a patient. An immunotoxin and an immune checkpoint inhibitor are administered to the patient. The immunotoxin comprises a single chain variable region antibody fused to a PE38 truncated Pseudomonas exotoxin. The single chain variable region antibody has CDR1, CDR2, and CDR3 regions as shown in SEQ ID NO: 6-11.

[0005] According to another aspect of the invention a kit is provided for treating a tumor. The kit comprises an immunotoxin and an immune checkpoint inhibitor. The immunotoxin comprises a single chain variable region antibody fused to a PE38 truncated Pseudomonas exotoxin, wherein the single chain variable region antibody has CDR1, CDR2, and CDR3 regions as shown in SEQ ID NO: 6-11;

[0006] These and other embodiments which will be apparent to those of skill in the art upon reading the specification provide the art with treatment methods, regimens, kits, and agents for treating glioblastomas and other tumors expressing epidermal growth factor (EGF) receptors i.e., EGFR and its mutants, e.g., EGFR variant III.

BRIEF DESCRIPTION OF THE DRAWINGS

[0007] FIG. 1. Possible mechanism for combination of regional cytotoxic therapy and immune checkpoint blockade. Cytotoxic therapy may engender targeted destruction of primary tumors and an `in situ vaccine` effect. Simultaneous blockade of immune checkpoint receptors PD1, PD-L1, TIM-3, LAG-3, and/or CSF-1R may augment the vaccine-mediated immunity. APCs: Antigen Presenting Cells; PD1: Programmed Death molecule 1; PD-L1: Programmed Death-Ligand 1; TIM-3: T cell Immunoglobulin and Mucin domain-3; LAG-3: Lymphocyte Activation Gene-3; CSF-1R: Colony Stimulating Factor 1 Receptor; NK/NKT: natural killer/natural killer T cells.

[0008] FIGS. 2A-2D Flow cytometry analyses of the CT-2A-mD2C7 cell line. (FIG. 2A) mouse H-2Kb (MHC class I) expression, (FIG. 2B) mouse H-2Db (MHC class I) expression, (FIG. 2C) mouse PD-L1 expression, and (FIG. 2D) D2C7-IT target antigen mEGFRvIII expression on CT-2A-mD2C7 cells.

[0009] FIG. 3 In vitro cytotoxicity of D2C7-IT on CT-2A-mD2C7 cells. WST1 assay was utilized to determine the in vitro cytotoxicity of D2C7-IT on CT-2A-mD2C7 cells. The IC.sub.50 of D2C7-IT on CT-2A-mD2C7 cells was 0.47 ng/ml (lower line) compared to that of the negative control immunotoxin, P588-IT (upper line, IC.sub.50>1000 ng/ml).

[0010] FIG. 4. Survival of C57BL/6 mice injected intracranially with CT2A-mD2C7 cells.

[0011] FIGS. 5A-5B. Phenotypic profile of the immune cells populating the CT2A-mD2C7 brain tumor microenvironment. FIG. 5A control brain and FIG. 5B CT2A-mD2C7 tumors. (F480lo+F480int+F480hi)=macrophages

[0012] FIG. 6. Experimental outline for D2C7-IT, BLZ945 combination therapy.

[0013] FIG. 7. Anti-tumor efficacy of D2C7-IT and BLZ945 combination therapy against the intracranial CT2A-mD2C7 glioma model.

[0014] FIGS. 8A-8B. In vivo efficacy of D2C7-IT+ (.alpha.CTLA-4 or .alpha.PD-1) mAb combination therapy in subcutaneous CT2A-mD2C7 glioma-bearing C57BL/6 immunocompetent mice. FIG. 8A shows all the treatment groups followed up to Day 35. FIG. 8B shows treatment groups G4-6 followed up to Day 62 after initial tumor inoculation. Note that 4 out of 10 and 5 out of 10 mice were cured only in treatment groups G5 and G6, respectively.

[0015] FIGS. 9A-9B. Tumor rechallenging studies for the cured mice in the combinational treatment groups (G5 and G6). FIG. 9A. Cured mice were first rechallenged with 10.sup.6 CT2A parental cells on the left flank on Day 72. FIG. 9B shows subsequent challenge with 3.times.10.sup.5 CT2A-mD2C7 cells in the brain on Day 126 after the initial CT2A-mD2C7 cell inoculation on the right flank. Tumors grew in all naive mice (G0), whereas no tumor grew in those cured mice (G5 and G6).

[0016] FIGS. 10A-10B. In vivo efficacy of D2C7-IT+ (.alpha.CTLA-4 or .alpha.PD-1) mAb combination therapy in bilateral subcutaneous CT2A-mD2C7 glioma-bearing C57BL/6 immunocompetent mice. FIG. 10A shows the tumor growth curve for the right (treated) tumors, which was similar to the previous one-side model. FIG. 10B shows the tumor growth curve for the left (untreated) tumors, which showed that the combinational therapy on the right side could also have an antitumor effect on the distant left tumor.

[0017] FIGS. 11A-11B. Left tumor volume among groups on Day 35 and Day 43 after the initial tumor inoculation. FIG. 11A. On Day 35, compared to treatment group G1, the left tumor growth was significantly delayed in all the other treatment groups, p<0.05 for treatment groups G2, G3, and G4; p<0.01 for treatment groups G5 and G6. FIG. 11B. On Day 43, the left tumor growth was significantly delayed by the combinational therapy compared to the D2C7-IT monotherapy on the right tumor, p<0.05.

DETAILED DESCRIPTION OF THE INVENTION

[0018] The inventors have developed targeted immunotoxins (IT), D2C7-(scdsFv)-PE38KDEL (D2C7-IT), by fusing the single chain variable fragment (scFv) from the D2C7 monoclonal antibody (mAb) with the Pseudomonas exotoxin A (PE), optionally fused to KDEL peptide. D2C7-IT reacts with both the wild-type epidermal growth factor receptor (EGFRwt) and the EGFR variant III (EGFRvIII), two proteins that are overexpressed in glioblastoma. The robust antitumor efficacy of D2C7-IT is mediated through PE in orthotopic glioma xenograft models in immunocompromised mice. In addition to direct tumor cell killing, the immunotoxin monotherapy induces a secondary antitumor immune response through the engagement of T cells. When the immunotoxin is administered in a combination regimen with an immune checkpoint inhibitor, improved and synergistic results are observed.

[0019] Other moieties which can be attached to the antibodies include those which provide additional beneficial properties. For example, a KDEL (lys-asp-glu-leu) tetra-peptide can be added at the carboxy-terminus of the protein to provide retention in the endoplasmic reticulum. Variants such as DKEL, RDEL, and KNEL which function similarly can also be used.

[0020] Tumors which can be treated are any that react with the D2C7 antibody. These include but are not limited to those in which at least one EGFRvIII allele is present. These may be found in breast, head and neck, brain, glioblastoma multiforme, astrocytoma, lung, or other tumors. It may be desirable to determine the presence of such an allele prior to therapy. This can be done using an oligonucleotide-based technique, such as PCR, or using an immunological technique, such as immunohistochemistry. It may be desirable to determine the amount, fraction, ratio, or percentage of cells in the tumor which express EGFR and/or EGFRvIII. The more cells which express EGFR on their surfaces, the more beneficial such antibody therapy is likely to be. Even tumors that express little to no EGFRvIII may be treated due to the ability of the antibody to bind to wild-type EGFR. Optionally, tumors may be tested prior to treatment for reactivity with D2C7 antibody. The immunotoxin itself could be used as an immunohistochemistry agent, before treatment, during treatment, or after treatment. A secondary reagent could be used with the immunotoxin for detection. It could, for example, recognize the Pseudomonas component of the immunotoxin.

[0021] Immunotoxins can be administered by any technique known in the art. Compartmental delivery may be desirable to avoid cytotoxicity for normal tissues that express EGFR. Suitable compartmental delivery methods include, but are not limited to delivery to the brain, delivery to a surgically created tumor resection cavity, delivery to a natural tumor cyst, and delivery to tumor parenchyma.

[0022] Tumors which can be treated by the method of the present invention are any which express epidermal growth factor receptor (EGFR), whether wild type, EGFRvIII, or other variants. Preferably the tumor expresses the receptor in amounts far exceeding expression by normal tissues. The mechanism of high level expression may be by genetic amplification, other alterations, whether genetic or epigenetic or post translational modifications. Exemplary tumors which can be treated include without limitation: malignant gliomas, breast cancer, head and neck squamous cell carcinoma, lung cancer.

[0023] Blockade of T cell immune checkpoint receptors, can be performed against any such targets, including but not limited to PD-1, PD-L1, TIM-3, LAG-3, CTLA-4, and CSF-1R and combinations of such checkpoint inhibitors. The immune checkpoint receptors may be on tumor cells or immune cells such as T cells, monocytes, microglia, and macrophages, without limitation. The agents which assert immune checkpoint blockade may be small chemical entities or polymers, antibodies, antibody fragments, single chain antibodies or other antibody constructs, including but not limited to bispecific antibodies and diabodies.

[0024] Immune checkpoint inhibitors which may be used according to the invention include any that disrupt the inhibitory interaction of cytotoxic T cells and tumor cells. These include but are not limited to anti-PD-1 antibody, anti-PD-L1 antibody, anti-CTLA4 antibody, anti-LAG-3 antibody, anti-TIM-3 antibody. The inhibitor need not be an antibody, but can be a small molecule or other polymer. If the inhibitor is an antibody it can be a polyclonal, monoclonal, fragment, single chain, or other antibody variant construct. Inhibitors may target any immune checkpoint known in the art, including but not limited to, CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, CSF-1R, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, IDO, A2aR, and the B-7 family of ligands. Combinations of inhibitors for a single target immune checkpoint or different inhibitors for different immune checkpoints may be used.

[0025] Examples of inhibitors of CSF-1R which may be used in the combination therapy with the immunotoxin include, without limitation, the following agents which are in clinical development: PLX3397, PLX486, RG7155, AMG820, ARRY-382, FPA008, IMC-CS4, JNJ-40346527, and MCS110.

[0026] The immune checkpoint inhibitor may be administered at the same time, before, or after the immunotoxin. Typically the two agents will be administered within 30, 28, 21, 14, 7, 4, 2, or 1 day(s) of each other. The agents may be given repeatedly, either serially or in a cycle of first and second agents. It may be advantageous but not necessary for the vaccine to be administered prior to the checkpoint inhibitor. But the reverse order may also be used. Priming of a cytotoxic T lymphocyte response by the immunotoxin may take from about 5 to about 14 days. Administration of the checkpoint inhibitor may beneficially be commenced during or after the priming period.

[0027] Immune checkpoint inhibitors may be administered by any appropriate means known in the art for the particular inhibitor. These include intravenous, oral, intraperitoneal, sublingual, intrathecal, intracavitary, intramuscularly, and subcutaneously.

[0028] Treatment regimens may include, in addition to delivery of the immunotoxin and immune checkpoint inhibitor(s), surgical removal of the tumor, surgical reduction of the tumor, chemotherapy, biological therapy, radiotherapy. These modalities are standard of care in many disease states, and the patient need not be denied the standard of care. The immunotoxin and immune checkpoint inhibitor(s) may be administered before, during, or after the standard of care. The immunotoxin and immune checkpoint inhibitor(s) may be administered after failure of the standard of care.

[0029] Kits may comprise, in a single divided or undivided container, the immunotoxin or its components or its encoding DNA and the immune checkpoint inhibitor or combination of immune checkpoint inhibitors. Storage stability may vary between the two agents so separate vessels may be used. Optionally one or both agents may be lyophilized or frozen.

[0030] Immunotoxins can directly kill cancer cells that express high levels of the targeted tumor antigen. Immunotoxin monotherapy can efficiently and directly destroy tumor cells expressing targeted epitopes, such as EGFRwt and/or its truncated variant, EGFRvIII, in malignant brain tumor xenograft models in immunocompromised mice. Immunotoxin therapy can induce a secondary anti-tumor immune response in a mouse model, which is different from the direct killing mechanism and needs the cooperation of the immune system. Since malignant brain tumors are always a heterogeneous mass, it is possible that some tumor cells can escape from the direct targeted attack of the immunotoxin therapy due to the lack of epitopes. For this reason, the secondary anti-tumor immune response stimulated by the immunotoxin may play an important role in eliminating those tumor cells not directly targeted.

[0031] Recently, several studies successfully demonstrated that tumor regression and significantly improved survival were achieved in murine glioma models by suppressing co-inhibitory molecules, such as CTLA4, CSF-1R, IDO, and PD1. Based on the promising preclinical data, several clinical trials have started to investigate the utilization of immune checkpoint inhibitors to treat malignant brain tumors, either as monotherapy or combinatorial therapy with other anti-tumor agents.

[0032] However, malignant gliomas, including glioblastomas, have relatively low mutation rates, which may generate fewer and subtle tumor antigens, leading to relatively poor basal immunogenicity compared to other tumor types that respond well to immunotherapies, for example, melanoma and NSCLC. Therefore, a combination of targeted cytotoxic immunotherapy and immune checkpoint inhibitors may provide a synergistic anti-tumor effect.

[0033] The ideal combinatorial therapy may have a lower dose of targeted cytotoxic immunotherapy to limit its side effects, and achieve long-term anti-tumor immunity. Immunotoxin therapy can efficiently and directly kill cancer cells that express high levels of the targeted antigen through its unique cytotoxic mechanism. Cancer cells destroyed by localized immunotoxin therapy release tumor antigens and/or other neoantigens. These antigens can then be presented by the APCs to host T cells in the local draining lymph nodes, which activate CTLs to migrate and eliminate the remaining or recurrent tumor cells expressing specific tumor antigens at the tumor site. Throughout this process, various co-inhibitory checkpoint pathways between T cells and APCs and/or between T cells and tumor cells can trigger different mechanisms to de-activate T cells, and to adjust the continuation and intensity of the anti-tumor immunity. Immune checkpoint inhibitors, such as anti-CTLA4 and anti-PD1 mAbs, can block these immunosuppressive pathways and therefore augment tumor cell death caused by lymphocytes activated by the targeted immunotoxin therapy.

[0034] We established a subcutaneous mouse CT2A-mD2C7 glioma model in C57BL/6 immunocompetent mice with six groups, in which the mice were treated by the control immunotoxin P588-IT or D2C7-IT, combined with .alpha.CTLA4 or .alpha.PD1 inhibitors after the tumor grew to a certain size. In this in vivo subcutaneous CT2A-mD2C7 glioma model, four doses of the low-dose D2C7-IT but not .alpha.CTLA4 or .alpha.PD1 monotherapy, and D2C7-IT+.alpha.CTLA-4 or .alpha.PD-1 combinatorial therapy generated a significant delay in tumor growth compared to the control immunotoxin P588-IT treatment groups (FIGS. 8A and 8B). Importantly, complete cures were only observed in D2C7-IT+.alpha.CTLA4 (n=4/10) and D2C7-IT+.alpha.PD-1 (n=5/10) combinatorial therapy groups (FIGS. 8A and 8B), although D2C7-IT monotherapy could also significantly delay the tumor growth. These results demonstrated that low doses of cytotoxic immunotoxin therapy can significantly delay the tumor growth but fail to cure the tumor-bearing mice. Combined with immune checkpoint inhibitors, cytotoxic immunotoxin therapy can increase the initial cure rate of low-dose immunotoxin therapy from zero to over 40%. Furthermore, all cured mice rejected the mD2C7-negative tumors primary subcutaneous rechallenge, whereas tumors grew in untreated naive mice, suggesting that the combinatorial treatment provided long lasting anti-tumor immunity that even extended to mD2C7-negative parental cells, as well (FIG. 9A). All nine cured mice then rejected the CT2A-mD2C7 secondary intracranial rechallenge in the brain, whereas tumors grew in untreated naive mice, indicating that the combinatorial treatment also provided long lasting anti-tumor immunity that extended to the remote immune-privileged CNS as well (FIG. 9B), in which the central memory T cells (T.sub.CM) might play an important role.

[0035] Subsequently, we established a bilateral subcutaneous mouse glioma model to investigate whether a localized high-dose immunotoxin treatment can provide a systemic anti-tumor effect on the tumors in the distal region, and whether the combination of immune checkpoint inhibitors can enhance this systemic anti-tumor immunity induced by the localized immunotoxin therapy. The D2C7-IT monotherapy, D2C7-IT+.alpha.CTLA4, and D2C7-IT+.alpha.PD1 combinatorial therapy led to significant growth delay of the right tumors (P<0.01), and cured 4/10, 6/10, and 5/10 right tumors, respectively (FIG. 10A). Interestingly, in the groups where the right tumors were treated by D2C7-IT or .alpha.CTLA-4 or .alpha.PD-1 monotherapy or D2C7-IT+(.alpha.CTLA-4 or .alpha.PD-1) combinatorial therapy, the left untreated tumors also grew much slower compared to the control group (FIGS. 10B and 11A), which indicates that a high dose of localized D2C7-IT monotherapy can achieve a similar anti-tumor immunity on the left untreated tumors compared to the systemic immune checkpoint inhibitor monotherapy. Furthermore, the combination therapy in the right tumors led to the most significantly delayed growth of the left untreated tumors in the mice (FIGS. 10B and 11B), which suggests that immune checkpoint inhibitors can enhance the anti-tumor immunity induced by the localized immunotoxin therapy to restrict the tumor growth in the remote area.

[0036] We have demonstrated that the intratumoral delivery of D2C7-IT induces secondary anti-tumor immunity, which destroys not only mD2C7-expressing tumor cells, but also tumor cells not expressing mD2C7 at the systemic level. A combination of D2C7 immunotoxin with immune checkpoint inhibitors can enhance this immunotoxin-induced anti-tumor immunity to achieve a synergistic long-term anti-tumor effect.

[0037] The above disclosure generally describes the present invention. All references disclosed herein are expressly incorporated by reference. A more complete understanding can be obtained by reference to the following specific examples which are provided herein for purposes of illustration only, and are not intended to limit the scope of the invention.

Example 1

[0038] We established a mouse glioma line, CT-2A-mD2C7, overexpressing the D2C7-IT antigen mouse EGFRvIII (mEGFRvIII). The reactivity and therapeutic efficacy of D2C7-IT against CT-2A-mD2C7 cells was determined by flow cytometry and in vitro cytotoxicity assays (WST1), respectively. CT-2A-mD2C7 cells were further analyzed for MHC class I and PD-L1 expression by flow cytometry. In vivo efficacy of D2C7-IT or .alpha.CTLA-4 or .alpha.PD-1 monotherapy or D2C7-IT+.alpha.CTLA-4 or D2C7-IT+.alpha.PD-1 combination therapy was evaluated in subcutaneous CT-2A-mD2C7 glioma-bearing C57BL/6 immunocompetent mice.

[0039] WST-1 is a reagent for measuring cell proliferation. It is used for the nonradioactive, spectrophotometric quantification of cell proliferation and viability in cell populations. The assay is based on the cleavage of the tetrazolium salt WST-1 to formazan by cellular mitochondrial dehydrogenases. Expansion in the number of viable cells results in an increase in the activity of the mitochondrial dehydrogenases, which in turn leads to increase in the amount of formazan dye formed. The formazan dye produced by viable cells can be quantified by measuring the absorbance at .lamda.=440 nm.

Example 2

[0040] Flow cytometry analysis confirmed the specific binding ability of D2C7 monoclonal antibody to the CT-2A-mD2C7 cells (FIG. 2D). Flow cytometry also demonstrated a high expression of both MHC class I molecules (FIGS. 2A and 2B) and PD-L1 (FIG. 2C) on tumor cell surface. D2C7-IT was highly cytotoxic (IC50=0.47 ng/mL) against CT-2A-mD2C7 cells in in vitro WST1 cytotoxicity assay (FIG. 3).

Example 3

[0041] Construction, expression, and purification of D2C7-(scdsFv)-PE38KDEL immunotoxin. The carboxyl terminus of the D2C7 V.sub.H domain was connected to the amino terminus of the V.sub.L domain by a 15-amino-acid peptide (Gly.sub.4Ser).sub.3 linker. In order to obtain a stable IT, it is essential to ensure that during renaturation V.sub.H is positioned near V.sub.L. This was achieved by mutating a single key residue in each chain to cysteine, for the stabilizing disulfide bond to form. On the basis of predictions using molecular modeling and empirical data with other dsFv-recombinant ITs, we chose one amino acid in each chain to mutate to cysteine. These are residues 44 in the framework region 2 (FR2) of V.sub.H and 100 in the FR4 of V.sub.L (according to the Kabat numbering). Thus, we prepared an Fv that contains both a peptide linker and a disulfide bond generated by cysteine residues that replace Ser44 of V.sub.H and Gly100 of V.sub.L. The D2C7 (scdsFv) PCR fragment was then fused to DNA for domains II and III of Pseudomonas exotoxin A. The version of Pseudomonas exotoxin A used here, PE38KDEL, has a modified C terminus which increases its intracellular retention, in turn enhancing its cytotoxicity. The D2C7-(scdsFv)-PE38KDEL was expressed in E. coli under the control of T7 promoter and harvested as inclusion bodies.

Example 4

[0042] Targeting tumor and tumor-associated macrophages for glioblastoma therapy: We evaluated the ability of D2C7-IT and BLZ945 combination treatment to function synergistically and produce an effective antitumor response in immunocompetent glioblastoma mouse models.

[0043] Intracranial growth curve for CT2A-mD2C7 in C57BL/6 mice: To determine the time course of CT2A-mD2C7 intracranial tumor growth, 3.times.10.sup.5 cells/3 .mu.l were implanted into 9 female C57BL/6 mice, and a survival curve was plotted (FIG. 4). The CT2A-mD2C7 survival curve demonstrated that 100% death occurred at day 25 post-tumor implantation. Based on our previous studies, post-tumor implantation day 8 was chosen as the optimal day to initiate D2C7-IT infusion.

[0044] Phenotypic profile of the immune cells populating the CT2A-mD2C7 brain tumor microenvironment: To characterize the immune cell phenotype of intracranial CT2A-mD2C7 tumors, C57BL/6 immunocompetent mice were implanted with 3.times.10.sup.5 tumor cells. The mice were followed to assess tumor development and were euthanized when they became moribund. Upon euthanization, the brains were harvested and the tumors were analyzed for infiltrating immune cells by flow cytometry. Cells isolated from naive C57BL/6 mice were used as the control. The primary cell types in the normal brain were microglia (80%), macrophages (F480lo+F480int+F480hi=11%), and T cells (5%) (FIG. 5A). However, there was a significant change in the percentage of microglia (8%), macrophages (F480lo+F480int+F480hi=63%), and T cells (19%) in tumor-bearing mice (FIG. 5B).

[0045] Anti-tumor efficacy of D2C7-IT and BLZ945 combination therapy against the intracranial CT2A-mD2C7 glioma model: The experimental outline for D2C7-IT+BLZ945 combination therapy against the CT2A-mD2C7 cell line is shown in FIG. 6. The in vivo efficacy of D2C7-IT or BLZ945 monotherapy or D2C7-IT+BLZ945 combination therapy was evaluated in intracranial CT2A-mD2C7 glioma-bearing C57BL/6 immunocompetent mice (FIG. 7). D2C7-IT (0.018 .mu.g total dose) was infused by an osmotic pump via convection-enhanced delivery (CED) for 72 hours from Day 8 to Day 11 post-tumor inoculation. BLZ945 (200 mg/kg) was delivered once daily through oral gavage on day 7 and on days 12-25. The survival curves for mice treated with the vehicle control and with BLZ945 monotherapy looked similar (Median Survival=22-24 days, FIG. 7). At a total dose of 0.018 .mu.g, D2C7-IT monotherapy extended the median survival to 42 days (FIG. 7). The median survival for the D2C7-IT+BLZ945 combination therapy group was 53 days (FIG. 7). Significantly, complete cures were observed only in the combination therapy group (2/6 mice). The preliminary data suggests that glioblastoma patients will benefit from D2C7-IT and BLZ945 combination therapy.

Example 5

In Vivo Efficacy of D2C7-IT+(Anti-CTLA4 or Anti-PD1) Inhibitors Combinatorial Therapy in a Subcutaneous (SC) CT2A-mD2C7 Glioma Model.

[0046] In previous pilot studies, we observed that the subcutaneous rechallenged mouse glioma allografts were rejected in those immunocompetent mice bearing SC mouse glioma allografts cured by the intratumoral (i.t.) immunotoxin therapy, suggesting that there can be a memory anti-tumor immune response following the SC immunotoxin therapy. This phenomenon was also reported in the SC melanoma mouse model treated by an immunotoxin targeting IL-13, in which CTLs played a major role in mediating this immunotoxin-induced anti-tumor response, although melanoma is a dramatically different type of tumor compared to malignant gliomas in the CNS. Therefore, it is necessary to establish appropriate mouse glioma models to investigate the secondary immune response, induced by immunotoxins, against glioblastomas, and to determine how to enhance this response by the combinatorial therapy of immune checkpoint inhibitors, such as anti-CTLA4 or anti-PD1 antibodies (.alpha.CTLA4 or .alpha.PD1), in order to achieve a long-lasting remission.

[0047] We established a subcutaneous mouse CT2A-mD2C7 glioma model in C57BL/6 immunocompetent mice with six groups, in which the mice were treated by the control immunotoxin P588-IT or D2C7-IT, combined with .alpha.CTLA4 or .alpha.PD1 inhibitors. In this in vivo subcutaneous CT2A-mD2C7 glioma model, four doses (every 3 days) of the D2C7-IT (low dose, 1.5 .mu.g per mouse per dose, i.t.) but not .alpha.CTLA4 (100 .mu.g per mouse per dose, intraperitoneal [i.p]) or .alpha.PD1 (250 .mu.g per mouse per dose, i.p.) monotherapy and D2C7-IT+.alpha.CTLA-4 or .alpha.PD-1 combinatorial therapy (immune checkpoint inhibitors administered on the next day after IT therapy) generated a significant delay in tumor growth compared to the control immunotoxin P588-IT treatment groups (P<0.01, FIG. 8A). Importantly, complete cures were only observed in D2C7-IT+.alpha.CTLA4 (n=4/10) and D2C7-IT+.alpha.PD-1 (n=5/10) combinatorial therapy groups (FIGS. 8A and 8B), although D2C7-IT monotherapy could also significantly delay the tumor growth.

Example 6

[0048] Tumor Rechallenging Studies on the Cured Mice from the D2C7-IT and Immune Checkpoint Inhibitors Combinatorial Treatment Groups

[0049] To determine whether those cured mice from D2C7-IT and immune checkpoint inhibitors combinatorial treatment groups can recall a protective anti-tumor memory immune response, on Day 72 after the initial tumor challenge, all nine cured mice were then first subcutaneously rechallenged (1.degree. SCR) with a dose of 10.sup.6 CT2A parental cells in the left flank. All these mice rejected the mD2C7-negative tumors, whereas tumors grew in all untreated naive mice, suggesting that the combinatorial treatment provided long lasting anti-tumor immunity that extended to mD2C7-negative parental cells as well (FIG. 9A).

[0050] To determine whether this protective anti-tumor immunity can protect the mice from the tumor rechallenging in a remote immune-privileged region, for example, the brain, all nine cured mice were then intracranially (IC) rechallenged (2.degree. ICR) for a second time on Day 126 (after the initial subcutaneous tumor challenge) with a dose of 3.times.10.sup.5 CT2A-mD2C7 cells in the brain. At the end of this study, all surviving mice were euthanized for brain histopathologic examination, which did not show tumors in the brains (data not shown). All these mice rejected the CT2A-mD2C7 intracranial (IC) rechallenge (2.degree. ICR), whereas tumors grew in all untreated naive mice, suggesting that the combinatorial treatment also provided long lasting anti-tumor immunity that extended to the remote immune-privileged CNS as well (FIG. 9B).

Example 7

In Vivo Efficacy of D2C7-IT+(Anti-CTLA4 or Anti-PD1) Inhibitors Combinatorial Therapy in a Bilateral Subcutaneous CT2A-mD2C7 Glioma Model.

[0051] In an in vivo bilateral subcutaneous CT2A-mD2C7 glioma model, tumor cells were inoculated in both sides of the flank simultaneously in C57BL/6 mice, with a high density (3.times.10.sup.6 cells) on the right side and a low density (10.sup.6 cells) on the left side. The larger tumors (on the right) were treated with four doses (every two days) of D2C7-IT or .alpha.CTLA4 or .alpha.PD1 monotherapy or D2C7-IT+.alpha.CTLA4 or D2C7-IT+.alpha.PD1 combination therapy (immune checkpoint inhibitors administered on the same day of immunotoxin therapy), while the left tumors were untreated. The D2C7-IT monotherapy (high dose, 4.5 .mu.g per mouse per dose, intratumoral), D2C7-IT+.alpha.CTLA4 (100 .mu.g per mouse per dose, intraperitoneal), and D2C7-IT+.alpha.PD1 (250 .mu.g per mouse per dose, intraperitoneal.) combinatorial therapies led to significant growth delays of the right tumor (P<0.01), which cured 4/10, 6/10, and 5/10 right tumors, respectively (FIG. 10A).

[0052] Interestingly, in the groups where the right tumors were treated with D2C7-IT or .alpha.CTLA-4 or .alpha.PD-1 monotherapy or D2C7-IT+(.alpha.CTLA-4 or .alpha.PD-1) combinatorial therapy, the left untreated tumors also grew much slower compared to the control group (FIGS. 10B and 11A), which indicates that a high dose of localized D2C7-IT monotherapy can achieve a similar anti-tumor immunity in the left untreated tumors compared to the systemic immune checkpoint inhibitor monotherapy. Furthermore, the D2C7-IT+(.alpha.CTLA-4 or .alpha.PD-1) combinatorial therapy in the right tumors led to the most significantly delayed growth of the left untreated tumors in the mice (FIGS. 10B and 11B), which suggests that immune checkpoint inhibitors can enhance the anti-tumor immunity induced by the immunotoxin to restrict the tumor growth in the remote area.

Sequence CWU 1

1

141120PRTArtificial Sequenceantibody construct, whole or portion 1Glu Val His Leu Gln Gln Ser Gly Pro Glu Leu Glu Lys Pro Gly Ala1 5 10 15Ser Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Ser Phe Thr Gly Tyr 20 25 30Asn Met Asn Trp Val Lys Gln Ser Asn Gly Lys Cys Leu Glu Trp Ile 35 40 45Gly Asn Ile Asp Pro Tyr Tyr Gly Asp Thr Asp Tyr Asp Gln Lys Phe 50 55 60Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Asn Thr Val Tyr65 70 75 80Met Gln Leu Gln Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys 85 90 95Ala Arg Gly Ala His Arg Asp Tyr Tyr Ala Met Asp Tyr Trp Gly Gln 100 105 110Gly Thr Ser Val Thr Val Ser Ser 115 120215PRTArtificial Sequenceantibody construct, whole or portion 2Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser1 5 10 153107PRTArtificial Sequenceantibody construct, whole or portion 3Asp Ile Gln Met Thr Gln Ser Pro Ala Ser Leu Ser Ala Ser Val Gly1 5 10 15Glu Thr Val Thr Ile Thr Cys Arg Thr Ser Glu Asn Ile Tyr Ile Tyr 20 25 30Leu Ala Trp Tyr Gln Gln Lys Gln Gly Lys Ser Pro Gln Leu Leu Val 35 40 45Tyr Asn Ala Lys Thr Leu Ala Glu Gly Val Pro Ser Arg Phe Ser Gly 50 55 60Ser Gly Ser Gly Thr Gln Phe Ser Leu Lys Ile Asn Gly Leu Gln Pro65 70 75 80Glu Asp Phe Gly Gly Tyr Tyr Cys Gln Gln His Tyr Gly Thr Pro Tyr 85 90 95Thr Phe Gly Cys Gly Thr Lys Leu Glu Lys Lys 100 1054351PRTArtificial Sequenceantibody construct, whole or portion 4Lys Ala Ser Gly Gly Pro Glu Gly Gly Ser Leu Ala Ala Leu Thr Ala1 5 10 15His Gln Ala Cys His Leu Pro Leu Glu Thr Phe Thr Arg His Arg Gln 20 25 30Pro Arg Gly Trp Glu Gln Leu Glu Gln Cys Gly Tyr Pro Val Gln Arg 35 40 45Leu Val Ala Leu Tyr Leu Ala Ala Arg Leu Ser Trp Asn Gln Val Asp 50 55 60Gln Val Ile Arg Asn Ala Leu Ala Ser Pro Gly Ser Gly Gly Asp Leu65 70 75 80Gly Glu Ala Ile Arg Glu Gln Pro Glu Gln Ala Arg Leu Ala Leu Thr 85 90 95Leu Ala Ala Ala Glu Ser Glu Arg Phe Val Arg Gln Gly Thr Gly Asn 100 105 110Asp Glu Ala Gly Ala Ala Asn Gly Pro Ala Asp Ser Gly Asp Ala Leu 115 120 125Leu Glu Arg Asn Tyr Pro Thr Gly Ala Glu Phe Leu Gly Asp Gly Gly 130 135 140Asp Val Ser Phe Ser Thr Arg Gly Thr Gln Asn Trp Thr Val Glu Arg145 150 155 160Leu Leu Gln Ala His Arg Gln Leu Glu Glu Arg Gly Tyr Val Phe Val 165 170 175Gly Tyr His Gly Thr Phe Leu Glu Ala Ala Gln Ser Ile Val Phe Gly 180 185 190Gly Val Arg Ala Arg Ser Gln Asp Leu Asp Ala Ile Trp Arg Gly Phe 195 200 205Tyr Ile Ala Gly Asp Pro Ala Leu Ala Tyr Gly Tyr Ala Gln Asp Gln 210 215 220Glu Pro Asp Ala Arg Gly Arg Ile Arg Asn Gly Ala Leu Leu Arg Val225 230 235 240Tyr Val Pro Arg Ser Ser Leu Pro Gly Phe Tyr Arg Thr Ser Leu Thr 245 250 255Leu Ala Ala Pro Glu Ala Ala Gly Glu Val Glu Arg Leu Ile Gly His 260 265 270Pro Leu Pro Leu Arg Leu Asp Ala Ile Thr Gly Pro Glu Glu Glu Gly 275 280 285Gly Arg Leu Glu Thr Ile Leu Gly Trp Pro Leu Ala Glu Arg Thr Val 290 295 300Val Ile Pro Ser Ala Ile Pro Thr Asp Pro Arg Asn Val Gly Gly Asp305 310 315 320Leu Asp Pro Ser Ser Ile Pro Asp Lys Glu Gln Ala Ile Ser Ala Leu 325 330 335Pro Asp Tyr Ala Ser Gln Pro Gly Lys Pro Pro Lys Asp Glu Leu 340 345 3505593PRTArtificial Sequenceantibody construct, whole or portion 5Glu Val His Leu Gln Gln Ser Gly Pro Glu Leu Glu Lys Pro Gly Ala1 5 10 15Ser Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Ser Phe Thr Gly Tyr 20 25 30Asn Met Asn Trp Val Lys Gln Ser Asn Gly Lys Cys Leu Glu Trp Ile 35 40 45Gly Asn Ile Asp Pro Tyr Tyr Gly Asp Thr Asp Tyr Asp Gln Lys Phe 50 55 60Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Asn Thr Val Tyr65 70 75 80Met Gln Leu Gln Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys 85 90 95Ala Arg Gly Ala His Arg Asp Tyr Tyr Ala Met Asp Tyr Trp Gly Gln 100 105 110Gly Thr Ser Val Thr Val Ser Ser Gly Gly Gly Gly Ser Gly Gly Gly 115 120 125Gly Ser Gly Gly Gly Gly Ser Asp Ile Gln Met Thr Gln Ser Pro Ala 130 135 140Ser Leu Ser Ala Ser Val Gly Glu Thr Val Thr Ile Thr Cys Arg Thr145 150 155 160Ser Glu Asn Ile Tyr Ile Tyr Leu Ala Trp Tyr Gln Gln Lys Gln Gly 165 170 175Lys Ser Pro Gln Leu Leu Val Tyr Asn Ala Lys Thr Leu Ala Glu Gly 180 185 190Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Gln Phe Ser Leu 195 200 205Lys Ile Asn Gly Leu Gln Pro Glu Asp Phe Gly Gly Tyr Tyr Cys Gln 210 215 220Gln His Tyr Gly Thr Pro Tyr Thr Phe Gly Cys Gly Thr Lys Leu Glu225 230 235 240Lys Lys Lys Ala Ser Gly Gly Pro Glu Gly Gly Ser Leu Ala Ala Leu 245 250 255Thr Ala His Gln Ala Cys His Leu Pro Leu Glu Thr Phe Thr Arg His 260 265 270Arg Gln Pro Arg Gly Trp Glu Gln Leu Glu Gln Cys Gly Tyr Pro Val 275 280 285Gln Arg Leu Val Ala Leu Tyr Leu Ala Ala Arg Leu Ser Trp Asn Gln 290 295 300Val Asp Gln Val Ile Arg Asn Ala Leu Ala Ser Pro Gly Ser Gly Gly305 310 315 320Asp Leu Gly Glu Ala Ile Arg Glu Gln Pro Glu Gln Ala Arg Leu Ala 325 330 335Leu Thr Leu Ala Ala Ala Glu Ser Glu Arg Phe Val Arg Gln Gly Thr 340 345 350Gly Asn Asp Glu Ala Gly Ala Ala Asn Gly Pro Ala Asp Ser Gly Asp 355 360 365Ala Leu Leu Glu Arg Asn Tyr Pro Thr Gly Ala Glu Phe Leu Gly Asp 370 375 380Gly Gly Asp Val Ser Phe Ser Thr Arg Gly Thr Gln Asn Trp Thr Val385 390 395 400Glu Arg Leu Leu Gln Ala His Arg Gln Leu Glu Glu Arg Gly Tyr Val 405 410 415Phe Val Gly Tyr His Gly Thr Phe Leu Glu Ala Ala Gln Ser Ile Val 420 425 430Phe Gly Gly Val Arg Ala Arg Ser Gln Asp Leu Asp Ala Ile Trp Arg 435 440 445Gly Phe Tyr Ile Ala Gly Asp Pro Ala Leu Ala Tyr Gly Tyr Ala Gln 450 455 460Asp Gln Glu Pro Asp Ala Arg Gly Arg Ile Arg Asn Gly Ala Leu Leu465 470 475 480Arg Val Tyr Val Pro Arg Ser Ser Leu Pro Gly Phe Tyr Arg Thr Ser 485 490 495Leu Thr Leu Ala Ala Pro Glu Ala Ala Gly Glu Val Glu Arg Leu Ile 500 505 510Gly His Pro Leu Pro Leu Arg Leu Asp Ala Ile Thr Gly Pro Glu Glu 515 520 525Glu Gly Gly Arg Leu Glu Thr Ile Leu Gly Trp Pro Leu Ala Glu Arg 530 535 540Thr Val Val Ile Pro Ser Ala Ile Pro Thr Asp Pro Arg Asn Val Gly545 550 555 560Gly Asp Leu Asp Pro Ser Ser Ile Pro Asp Lys Glu Gln Ala Ile Ser 565 570 575Ala Leu Pro Asp Tyr Ala Ser Gln Pro Gly Lys Pro Pro Lys Asp Glu 580 585 590Leu65PRTArtificial Sequenceantibody construct, whole or portion 6Gly Tyr Asn Met Asn1 5717PRTArtificial Sequenceantibody construct, whole or portion 7Asn Ile Asp Pro Tyr Tyr Gly Asp Thr Asp Tyr Asp Gln Lys Phe Lys1 5 10 15Gly811PRTArtificial Sequenceantibody construct, whole or portion 8Gly Ala His Arg Asp Tyr Tyr Ala Met Asp Tyr1 5 10911PRTArtificial Sequenceantibody construct, whole or portion 9Arg Thr Ser Glu Asn Ile Tyr Ile Tyr Leu Ala1 5 10107PRTArtificial Sequenceantibody construct, whole or portion 10Asn Ala Lys Thr Leu Ala Glu1 5119PRTArtificial Sequenceantibody construct, whole or portion 11Gln Gln His Tyr Gly Thr Pro Tyr Thr1 5121779DNAArtificial Sequenceantibody construct, whole or portion 12gaggtccacc tgcagcagtc tggacctgag ctggagaagc ctggcgcttc agtgaagata 60tcctgcaagg cttctggtta ctcattcact ggctacaaca tgaactgggt gaagcagagc 120aatggcaagt gccttgagtg gattggaaat attgatcctt actatggtga tactgactac 180gaccagaagt tcaagggcaa ggccacattg actgcagaca aatcctccaa cacagtctac 240atgcagctcc agagcctgac atctgaggac tctgcagtct attactgtgc aagaggggcc 300catagggatt actatgctat ggactactgg ggtcaaggga cctcagtcac cgtctcctca 360ggtggtggcg gttcaggcgg aggtggctct ggcggtggcg gatcggacat ccagatgact 420cagtctccag cctccctatc tgcatctgtg ggagaaactg tcaccatcac atgtcgaaca 480agtgagaata tttacattta tttagcatgg tatcagcaga aacagggaaa atctcctcag 540ctcctggtct ataatgcaaa aaccttagca gaaggtgtgc catcaaggtt cagtggcagt 600gggtcaggca cacagttttc tctgaagatc aacggcctgc agcctgaaga ttttgggggt 660tattactgtc aacagcatta tggcactccg tacacgttcg gatgcgggac caagctggaa 720aaaaaaaaag cttccggagg tcccgagggc ggcagcctgg ccgcgctgac cgcgcaccag 780gcttgccacc tgccgctgga gactttcacc cgtcatcgcc agccgcgcgg ctgggaacaa 840ctggagcagt gcggctatcc ggtgcagcgg ctggtcgccc tctacctggc ggcgcggctg 900tcgtggaacc aggtcgacca ggtgatccgc aacgccctgg ccagccccgg cagcggcggc 960gacctgggcg aagcgatccg cgagcagccg gagcaagccc gtctggccct gaccctggcc 1020gccgccgaga gcgagcgctt cgtccggcag ggcaccggca acgacgaggc cggcgcggcc 1080aacggcccgg cggacagcgg cgacgccctg ctggagcgca actatcccac tggcgcggag 1140ttcctcggcg acggcggcga cgtcagcttc agcacccgcg gcacgcagaa ctggacggtg 1200gagcggctgc tccaggcgca ccgccaactg gaggagcgcg gctatgtgtt cgtcggctac 1260cacggcacct tcctcgaagc ggcgcaaagc atcgtcttcg gcggggtgcg cgcgcgcagc 1320caggacctcg acgcgatctg gcgcggtttc tatatcgccg gcgatccggc gctggcctac 1380ggctacgccc aggaccagga acccgacgca cgcggccgga tccgcaacgg tgccctgctg 1440cgggtctatg tgccgcgctc gagcctgccg ggcttctacc gcaccagcct gaccctggcc 1500gcgccggagg cggcgggcga ggtcgaacgg ctgatcggcc atccgctgcc gctgcgcctg 1560gacgccatca ccggccccga ggaggaaggc gggcgcctgg agaccattct cggctggccg 1620ctggccgagc gcaccgtggt gattccctcg gcgatcccca ccgacccgcg caacgtcggc 1680ggcgacctcg acccgtccag catccccgac aaggaacagg cgatcagcgc cctgccggac 1740tacgccagcc agcccggcaa accgccgaaa gacgagctc 177913726DNAArtificial Sequenceantibody construct, whole or portion 13gaggtccacc tgcagcagtc tggacctgag ctggagaagc ctggcgcttc agtgaagata 60tcctgcaagg cttctggtta ctcattcact ggctacaaca tgaactgggt gaagcagagc 120aatggcaagt gccttgagtg gattggaaat attgatcctt actatggtga tactgactac 180gaccagaagt tcaagggcaa ggccacattg actgcagaca aatcctccaa cacagtctac 240atgcagctcc agagcctgac atctgaggac tctgcagtct attactgtgc aagaggggcc 300catagggatt actatgctat ggactactgg ggtcaaggga cctcagtcac cgtctcctca 360ggtggtggcg gttcaggcgg aggtggctct ggcggtggcg gatcggacat ccagatgact 420cagtctccag cctccctatc tgcatctgtg ggagaaactg tcaccatcac atgtcgaaca 480agtgagaata tttacattta tttagcatgg tatcagcaga aacagggaaa atctcctcag 540ctcctggtct ataatgcaaa aaccttagca gaaggtgtgc catcaaggtt cagtggcagt 600gggtcaggca cacagttttc tctgaagatc aacggcctgc agcctgaaga ttttgggggt 660tattactgtc aacagcatta tggcactccg tacacgttcg gatgcgggac caagctggaa 720aaaaaa 726141053DNAArtificial Sequenceantibody construct, whole or portion 14aaagcttccg gaggtcccga gggcggcagc ctggccgcgc tgaccgcgca ccaggcttgc 60cacctgccgc tggagacttt cacccgtcat cgccagccgc gcggctggga acaactggag 120cagtgcggct atccggtgca gcggctggtc gccctctacc tggcggcgcg gctgtcgtgg 180aaccaggtcg accaggtgat ccgcaacgcc ctggccagcc ccggcagcgg cggcgacctg 240ggcgaagcga tccgcgagca gccggagcaa gcccgtctgg ccctgaccct ggccgccgcc 300gagagcgagc gcttcgtccg gcagggcacc ggcaacgacg aggccggcgc ggccaacggc 360ccggcggaca gcggcgacgc cctgctggag cgcaactatc ccactggcgc ggagttcctc 420ggcgacggcg gcgacgtcag cttcagcacc cgcggcacgc agaactggac ggtggagcgg 480ctgctccagg cgcaccgcca actggaggag cgcggctatg tgttcgtcgg ctaccacggc 540accttcctcg aagcggcgca aagcatcgtc ttcggcgggg tgcgcgcgcg cagccaggac 600ctcgacgcga tctggcgcgg tttctatatc gccggcgatc cggcgctggc ctacggctac 660gcccaggacc aggaacccga cgcacgcggc cggatccgca acggtgccct gctgcgggtc 720tatgtgccgc gctcgagcct gccgggcttc taccgcacca gcctgaccct ggccgcgccg 780gaggcggcgg gcgaggtcga acggctgatc ggccatccgc tgccgctgcg cctggacgcc 840atcaccggcc ccgaggagga aggcgggcgc ctggagacca ttctcggctg gccgctggcc 900gagcgcaccg tggtgattcc ctcggcgatc cccaccgacc cgcgcaacgt cggcggcgac 960ctcgacccgt ccagcatccc cgacaaggaa caggcgatca gcgccctgcc ggactacgcc 1020agccagcccg gcaaaccgcc gaaagacgag ctc 1053

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed