Targeted combination therapy

文档序号:1570292 发布日期:2020-01-24 浏览:34次 中文

阅读说明:本技术 靶向组合治疗 (Targeted combination therapy ) 是由 伊丽莎白·德洛斯皮诺斯 约翰·托德·席勒 罗达·C·基尼斯 于 2018-04-11 设计创作,主要内容包括:本公开内容的一些实施方案涉及使用免疫治疗剂和肿瘤靶向性病毒衣壳蛋白装配体的组合来治疗肿瘤的方法和组合物,所述肿瘤靶向性病毒衣壳蛋白装配体包含与病毒衣壳蛋白缀合的抗癌分子。(Some embodiments of the present disclosure relate to methods and compositions for treating tumors using a combination of immunotherapeutic agents and tumor-targeting viral capsid protein assemblies comprising an anti-cancer molecule conjugated to a viral capsid protein.)

1. A method, comprising:

(a) administering to a subject having a tumor a composition comprising a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of said assembly; and

(b) administering to the subject having the tumor a composition comprising an immune checkpoint inhibitor.

2. The method of claim 1, wherein the viral capsid protein is a human papilloma virus capsid protein.

3. The method of claim 1, wherein the viral capsid protein is a non-human papilloma virus capsid protein.

4. The method of claim 3, wherein the non-human papillomavirus capsid protein is a bovine papillomavirus capsid protein or a rabbit papillomavirus.

5. The method of claim 1, wherein the viral capsid protein comprises a papillomavirus L1 capsid protein or a combination of a papillomavirus L1 capsid protein and a papillomavirus L2 capsid protein.

6. The method of claim 1, wherein the tumor-targeting viral capsid protein assembly is a capsomer or a virus-like particle.

7. The method of claim 1, wherein the tumor-targeting viral capsid protein assembly comprises from 10 to 1000 photosensitive molecules, optionally wherein the tumor-targeting viral capsid protein assembly comprises from 200 to 500 photosensitive molecules.

8. The method of claim 1, wherein the photosensitive molecule comprises a fluorescent dye, an Infrared (IR) dye, a Near Infrared (NIR) dye, a porphyrin, a chlorophyll, a chlorin, a phthalocyanine, a bacteriochlorin, a deuteroporphyrin, a porfimer sodium, a verteporfin, a temoporfin, a talaporfin, a methylene blue, an aminolevulinic acid, or a combination thereof.

9. The method of claim 1, wherein the tumor-targeting viral capsid protein assembly is a virus-like particle comprising 72 capsomeres assembled from variant or modified HPV16/31L1 capsid protein and wild-type HPV L2 capsid protein, and wherein the photosensitive molecule comprises IRdye700 DX.

10. The method of claim 1, wherein the immune checkpoint inhibitor inhibits binding of CTLA-4, PD-1, PD-L1, TIM3, LAG3, B7-H3, B7-H4, BTLA, GAL9, Chk1, or A2aR to a respective binding partner.

11. The method of claim 10, wherein the immune checkpoint inhibitor is an antibody.

12. The method of claim 11, wherein the antibody is a monoclonal antibody.

13. The method of claim 1, wherein the antibody is selected from the group consisting of an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM 3 antibody, an anti-LAG 3 antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-BTLA antibody, an anti-GAL 9 antibody, an anti-Chk 1 antibody, an anti-A2 aR antibody, and a combination of any two or more of the foregoing antibodies.

14. The method of claim 13, wherein the antibody is selected from the group consisting of pembrolizumab, nivolumab, ipilimumab, and any combination of two or more of the foregoing antibodies.

15. The method of claim 1, wherein the tumor-targeting viral capsid protein assembly and immune checkpoint inhibitor are administered simultaneously.

16. The method of claim 1, wherein the tumor-targeting viral capsid protein assembly and immune checkpoint inhibitor are administered sequentially.

17. The method of claim 16, wherein the tumor-targeting viral capsid protein assemblies are administered to the subject prior to administration of the immune checkpoint inhibitor to the subject.

18. The method of claim 16, wherein the tumor-targeting viral capsid protein assemblies are administered locally to a tumor or tumor lesion, and wherein the immune checkpoint inhibitor is administered systemically.

19. The method of claim 1, wherein the tumor-targeting viral capsid protein assemblies and/or immune checkpoint inhibitors are administered by injection, topically or by implantation.

20. The method of claim 19, wherein the injection is intralesional injection, subcutaneous injection, intravitreal injection, suprachoroidal injection, intraperitoneal injection, intraarterial injection, intrahepatic injection, intravesical injection, or any combination thereof.

21. The method of claim 1, further comprising activating the photosensitive molecules using an infrared laser, a near-infrared laser, or an ultraviolet laser.

22. The method of claim 1, wherein the tumor is cancerous.

23. The method of claim 23, wherein the tumor is metastatic.

24. The method of claim 1, wherein the tumor is melanoma, carcinoma, sarcoma, or lymphoma.

25. The method of claim 1, wherein the tumor has a lesion that is treatable with an infrared laser.

26. The method of claim 25, wherein the tumor is a merkel cell carcinoma, squamous cell carcinoma, basal cell carcinoma, metastatic breast cancer, cutaneous T-cell lymphoma or sarcoma.

27. The method of claim 1, wherein the tumor is a primary tumor and has metastasized.

28. The method of claim 1, wherein the tumor is located in and/or has metastasized to: liver, bladder, eye, head, neck, cervix, larynx, skin, lung, pleura, pancreas, stomach, esophagus, colon, breast, ovary, prostate, testis, brain, meninges, kidney.

29. The method of claim 1, wherein the subject is a human.

30. A method comprising administering an immune checkpoint inhibitor to a subject having a tumor undergoing treatment with a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of said assembly.

31. A composition comprising an immune checkpoint inhibitor and a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of said assembly.

32. The composition of claim 31, wherein the viral capsid protein is a human papilloma virus capsid protein.

33. The composition of claim 31, wherein the viral capsid protein is a non-human papilloma virus capsid protein.

34. The composition of claim 33, wherein the non-human papillomavirus capsid protein is a bovine papillomavirus capsid protein or a rabbit papillomavirus.

35. The composition of claim 31, wherein the viral capsid protein comprises a papillomavirus L1 capsid protein or a combination of a papillomavirus L1 capsid protein and a papillomavirus L2 capsid protein.

36. The composition of claim 31, wherein the tumor-targeting viral capsid protein assembly is a capsomer or a virus-like particle.

37. The composition of claim 31, wherein the tumor-targeting viral capsid protein assembly comprises 10 to 1000 photosensitive molecules.

38. The composition of claim 31, wherein the photosensitive molecule comprises a fluorescent dye, an Infrared (IR) dye, a Near Infrared (NIR) dye, a porphyrin, a chlorophyll, a chlorin, a phthalocyanine, a bacteriochlorin, a deuteroporphyrin, a porfimer sodium, a verteporfin, a temoporfin, a talaporfin, a methylene blue, an aminolevulinic acid, or a combination thereof.

39. The composition of claim 31, wherein the tumor-targeting viral capsid protein assembly is a virus-like particle comprising 72 capsomeres assembled from variant or modified HPV16/31L1 capsid protein and wild-type HPV L2 capsid protein, and wherein the photosensitive molecule comprises IRdye700 DX.

40. The composition of claim 31, wherein the immune checkpoint inhibitor inhibits binding of CTLA-4, PD-1, PD-L1, TIM3, LAG3, B7-H3, B7-H4, BTLA, GAL9, Chk1, or A2aR to a respective binding partner.

41. The composition of claim 40, wherein the immune checkpoint inhibitor is an antibody.

42. The composition of claim 41, wherein the antibody is a monoclonal antibody.

43. The composition of claim 31, wherein the antibody is selected from the group consisting of an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM 3 antibody, an anti-LAG 3 antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-BTLA antibody, an anti-GAL 9 antibody, an anti-Chk 1 antibody, an anti-A2 aR antibody, and a combination of any two or more of the foregoing antibodies.

44. The composition of claim 43, wherein the antibody is selected from the group consisting of pembrolizumab, nivolumab, ipilimumab, and any combination of two or more of the foregoing antibodies.

Background

Although there are many treatments available for cancer, many forms of cancer remain incurable, untreatable, or resistant to standard therapies. Traditional cancer treatment methods are often associated with serious side effects due to cytotoxicity caused by anticancer drugs in normal/healthy cells.

Disclosure of Invention

The present disclosure provides methods and compositions for selectively targeting and killing tumor cells while simultaneously presenting tumor antigens to the immune system and synergistically generating anti-tumor immunity, thereby providing a combination therapy for the treatment and prevention of cancer. Unexpectedly, experimental data show that tumor-targeted photosensitive molecules that become cytotoxic after laser activation trigger a tumor antigen-specific immune response. To enhance this immune response against cancer cells, an immune checkpoint inhibitor may be administered simultaneously (or sequentially) with the tumor-targeting photosensitive molecule.

Thus, in some embodiments, the methods provided herein comprise: (a) administering to a subject having a tumor a composition comprising a tumor-targeting viral capsid protein assembly (assembly) comprising a photosensitive molecule conjugated to a viral capsid protein of the assembly; and (b) administering a composition comprising an immune checkpoint inhibitor to a subject having a tumor.

In some embodiments, an immune checkpoint inhibitor is administered to a subject having a tumor undergoing treatment with a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of the assembly.

Also provided herein are compositions comprising an immune checkpoint inhibitor and a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of the assembly.

The above summary is intended to illustrate, in a non-limiting manner, some embodiments, advantages, features, and uses of the technology disclosed herein. Other embodiments, advantages, features, and uses of the technology disclosed herein will be apparent from the detailed description, drawings, examples, and claims.

Drawings

FIG. 1 is a schematic diagram depicting the mechanism of action of a VLP conjugate of the present disclosure comprising a conjugate having a photosensitive molecule: (

Figure BDA0002311164830000021

700DX) to primary amines on the surface of the VLPs. The VLP conjugate is inactive in the absence of irradiation. The VLP conjugates preferentially target tumor cells. Infrared irradiation activates the VLP conjugate at 689 nm. The L1 protein of the viral capsid specifically binds to Heparan Sulfate Proteoglycans (HSPGs) on the tumor surface in a multivalent manner. The activated drug then disrupts the tumor cell membrane, resulting in pro-immunogenic tumor cell damage and necrosis.

FIG. 2 is a graph showing survival curves of mice in the TC-1 model. After forming about 50mm3After the tumor (day 0), mice were injected with VLP conjugate or Phosphate Buffered Saline (PBS) and then treated with light after 12 and 24 hours. The protocol was repeated on day 3.

FIG. 3 is a graph showing survival curves of mice in the TC-1 model. Is formed to be about 100 to 300mm3After the tumor (day 0), mice were injected with VLP conjugate or PBS and then treated with light after 12 hours. The protocol was repeated on day 3. Blood was drawn on days 0 and 17 to measure tumor specific T cell responses.

FIG. 4 shows the tumor antigen specific CD8 detected in the blood of experimental mice on day 17+T cells. Tumor volume, HPV 16E 7 tetramer is shown+CD8+Gating and HPV 16E 7 peptide restimulation-IFN-gamma, CD8+And a door is arranged.

FIG. 5 illustrates that light treatment alone does not result in tumor antigen specific CD8+T cells. Tumor volume, HPV 16E 7 tetramer is shown+CD8+Gating, and HPV 16E 7 peptide restimulation-IFN-gamma, CD8+And a door is arranged.

Figure 6 is a schematic illustrating the combination of tumor targeting drug conjugates activated with NIR and checkpoint inhibitors to prevent tumor growth and recurrence.

FIG. 7 is a schematic diagram of the experiment carried out in example 4.

Detailed Description

In order to deliver cancer/tumor therapeutic drugs specifically to tumors, certain viral capsid proteins can be assembled and/or chemically modified to carry therapeutic molecules without losing their tumor targeting ability or structural stability. These "viral capsid protein assemblies" can selectively bind to the surface of tumor cells, e.g., by binding to Heparan Sulfate Proteoglycans (HSPGs) on the surface of tumor cells, and enter the tumor cells, where therapeutic molecules (e.g., photosensitive molecules) can produce targeted cytotoxic effects and/or anti-tumor immunity, resulting in tumor cell death and prevention of tumor recurrence. The use of such virus-like particles and assemblies in the treatment of cancer is described, for example, in PCT application publication WO2015042325 filed on 9, 18, 2014, which is incorporated herein by reference in its entirety.

Some aspects of the present disclosure are based, at least in part, on unexpected results demonstrating that photosensitive molecules delivered to tumor cells via tumor-targeting viral capsid protein assemblies produce anti-tumor immunity. Tumor cells undergoing proammunogenic cell death release immunogenic factors, thereby stimulating Antigen Presenting Cells (APCs) to take up tumor antigens, process them, and induce tumor antigen-specific T cell immune responses against tumors. Such tumor antigen specific T cell responses, combined with therapeutic necrotic effects, show great potential for the selective and efficient destruction of tumor cells without affecting normal cells, resulting in anti-tumor immunity and the prevention of tumor recurrence.

Tumor cells often use the mechanisms adopted by the immune system to prevent self-attack. Cancer immunotherapeutics have been developed to reduce or eliminate the ability of tumor cells to evade attack by the immune system. The use of such immunotherapeutic agents for the treatment of cancer has been described, for example, in U.S. patent application publications US20160024469, US20130202645, US20100189641, U.S. patents US5478556, US5290551 and US5126129, the entire contents of which are incorporated herein by reference. In some embodiments, the immunotherapeutic agent is an immune checkpoint inhibitor. Thus, the present disclosure encompasses compositions and methods for combination treatment of cancer/tumors that take advantage of the unexpected synergistic effects produced by targeting and activating cytotoxic photosensitive molecules in tumor cells and administering immune checkpoint inhibitors (or other immunotherapeutic agents) simultaneously or sequentially. Such compositions and methods are more effective in controlling tumor growth, inducing anti-tumor immunity, and preventing tumor recurrence when compared to traditional chemotherapeutic methods and tumor immunotherapy strategies.

Accordingly, some aspects of the present disclosure provide compositions comprising tumor-targeting viral capsid protein assemblies comprising photosensitive molecules conjugated to viral capsid proteins of the assemblies.

A "viral capsid protein assembly" is any particle or aggregate formed from viral capsid proteins. Examples of viral capsid protein assemblies include, but are not limited to: capsomers (capsids) and virus-like nanoparticles, as well as other viral pseudoviruses and viral nanoparticles. The viral capsid protein assembly is considered "tumor-targeted" in that it binds to tumor (e.g., cancerous) cells and not to non-tumor (e.g., non-cancerous, other normal, healthy) cells. In some embodiments, the tumor-targeting viral capsid protein assembly is a virus-like particle (VLP) having 72 capsomeric structures (e.g., comprising L1 or L1/L2 viral capsid proteins).

In some embodiments, the viral capsid protein assembly is a Human Papilloma Virus (HPV) capsid protein assembly (e.g., from type 16, type 31 or a modified type comprising amino acids from type 16 and from type 31). In some embodiments, the viral capsid protein assembly is a non-Human Papillomavirus Virus (HPV) capsid protein assembly. Examples of non-human papillomavirus virus capsid protein assemblies include, but are not limited to, bovine papillomavirus, cotton rabbit papillomavirus, rhesus papilloma virus, and murine papilloma virus.

The viral capsid protein assembly comprises viral capsid proteins. The "capsid protein" is a protein monomer. Capsid proteins can assemble together to form capsomeres (e.g., pentamers of capsid proteins). A "capsomere" is a subunit of the viral capsid, which is the outer shell of proteins that protect the genetic material of a virus, such as Human Papilloma Virus (HPV). However, it is understood that, in the context of the present disclosure, capsomeres can be used as delivery vehicles (tumor-targeted delivery vehicles) independently of the capsid. Capsid proteins of the present disclosure include papillomavirus L1 capsid protein, papillomavirus L2 capsid protein, and variants thereof (e.g., variants having a reduced or modified ability to induce infection-inhibiting antibodies). In some embodiments, the viral capsid protein assemblies comprise only L1 capsid proteins, while in other embodiments, the viral capsid protein assemblies comprise a combination of L1 and L2 capsid proteins. As used herein, "outer capsid protein" refers to a capsid protein exposed on the surface of a VLP.

Virus-like particles or VLPs as used herein refer to organized capsid-like structures (e.g., roughly spherical or cylindrical) comprising self-assembled ordered arrays of L1 or L1 and L2, capsomeres, and no viral genome. In some embodiments, the virus-like particle is morphologically and antigenically similar to an authentic virus particle, but it lacks viral genetic material (e.g., viral nucleic acid), rendering the particle non-infectious.

In some embodiments, tumor-targeting viral capsid protein assemblies can be modified to reduce immunogenicity against the particle itself, i.e., to reduce induction of neutralizing antibodies against the assemblies. The viral capsid protein assemblies can be assembled, for example, from capsomeres containing capsid protein variants with modified immunogenicity. Capsid protein variants having "modified immunogenicity" are capsid protein variants that are naturally or synthetically modified (e.g., mutated, substituted, deleted, pegylated, or inserted) in amino acids to reduce or prevent recognition of the capsid protein by pre-existing (e.g., endogenous) viral serotype-specific antibodies. The capsid protein variant may be a Human Papillomavirus (HPV) L1 variant, a non-human papillomavirus L1 variant, or a papillomavirus L1 variant based on a combination of amino acids from different HPV serotypes. For example, an L1 variant with modified immunogenicity may be a recombinant protein described in international publication No. wo2010120266, filed 24/7/2009, the entire content of which is incorporated herein by reference. In some embodiments, the viral capsid protein assembly comprises an L1 variant (HPV16/31L1 capsid protein variant) with modified immunogenicity.

Tumor-targeting viral capsid protein assemblies of the present disclosure can specifically target cancer cells, and such specificity is mediated at least in part by binding of L1 protein in the viral capsid protein assemblies to Heparan Sulfate Proteoglycans (HSPGs) on the surface of tumor cells. This process is similar to the process by which a virus attaches and infects its host cells. Tumor-targeted viral capsid protein assemblies do not bind or pseudotarget (pseudo-target) intact epithelial cells and are therefore excellent tools for specific delivery of anticancer drugs to tumors without affecting healthy or normal cells.

The tumor-targeting viral capsid protein assemblies of the present disclosure comprise capsid proteins conjugated to molecules with anti-cancer activity (referred to herein as "anti-cancer molecules"). Such anti-cancer molecules are capable of producing targeted cytotoxic effects on tumor cells and/or in some cases may induce immunity against tumor cells. Non-limiting examples of molecules of the present disclosure include: bortezomib, imatinib, celecoxib, afatinib (Gilotrif), Aleninib (Alecensa), axinib (Inlyta), belinostat (Beleodaq), bortezomib (Velcade), bosutinib (Bosulif), Cabozantinib (Cabozantinib) (Cometriq), carfilzomib (Kyprolis), ceritinib (Zykadia), Cobimetinib (Cotemetiib) (Cotellic), crizotinib (Xalkori), Darafenib (Tafinalratin), Dasatinib (Sprycel), erlotinib (Tarceva), everolimus (Afiniborrelib), gefitinib (Tyranostimul), Icarinib (Zyigesbanib), erlotinib (Tyrosavi), Equisetimib (Oscilaria), Equisetimib (Osislandisib) (Osislandic), Icarinib (Osislandicib) (Osislandic), Icelatinib (Icelatinib) (Osisabib), Icelatinib (Osisabib) (Oscil (Osislandia), Icelatinib (Icelatinib) (Osisabib) (Osisb), Icelatinib (Oscil (Icelatinib) (Osisb), Icelatinib (Icelatinib) (Osisb) (Oscil (Icelosib) (Oscil), Icelosib) (Oscil (Paiparib) (Osisb) (Oscil (Icelabib) (Oscil), Icelabib) (Oscil, Ponatinib (Ponatinib) (iclusisig), regorafenib (Stivarga), ruxolitinib (Jakafi), Sipuleucel-t (provenge), sonedgib (Sonidegib) (Odomzo), sorafenib (Nexavar), sitaxolimus (torsel), Tofacitinib (Tofacitinib) (Xeljanz), tremetinib (meist), vandetanib (caprella), vemurafenib (Zelboraf), vismodegib (eridge), and vorinostat (zonza). Any anti-cancer molecule known in the art may be used in accordance with the present disclosure.

In some embodiments, the anti-cancer molecule is a photoactive molecule or a photosensitizer. A "photoactive molecule" or "photosensitizer" is a compound that can be excited to an excited state upon absorption of light. Such species rapidly attack any organic compounds they encounter and are therefore highly cytotoxic. In some embodiments, the activated photosensitive molecules re-emit light upon photoexcitation (e.g., fluorophores). In some embodiments, upon photoexcitation, the activated photosensitive molecules may become toxic, or toxic molecules may be generated. For example, a class of photosensitive molecules can be excited to an excited state upon absorption of light and cross with an oxygen generating system to produce singlet oxygen.

Unexpectedly, tumor-targeting viral capsid protein assemblies comprise photosensitive molecules conjugated to viral capsid proteins, which upon activation by NIR light induce not only tumor necrosis but also pro-immunogenic cell death of tumors and activate a durable tumor antigen specific T-cell response against tumors. In this way, the immune system is activated to attack the tumor, thereby enhancing local (regional) and distance treatment efficacy. Unexpectedly, this mechanism of action not only reduces the size of existing tumors, i.e., inhibits tumor growth, but also prevents mice from developing new tumors after challenge, i.e., prevents tumor recurrence.

Examples of photoactive molecules used in accordance with the present disclosure include, but are not limited to, fluorescent dyes, infrared dyes, near-infrared dyes, porphyrin molecules, and chlorophyll molecules.

Examples of fluorescent dyes for use in accordance with the present disclosure include, but are not limited to: acridine orange (acridineoorange), acridine yellow (acridine yellow), Alexa Fluor, 7-amino actinomycin D, 8-anilinonaphthalene a-1-sulfonic acid, ATTO dyes, auramine-rhodamine dyes (auramine-rhodomine stain), benzanthrone, bimane, 9, 10-bis (phenylethynyl) anthracene, 5, 12-bis (phenylethynyl) tetracene, bisbenzimide, black light lacquer, calcein (calcein), carboxyfluorescein diacetate succinimidyl ester, carboxyfluorescein succinimidyl ester, 1-chloro-9, 10-bis (phenylethynyl) anthracene, 2-chloro-9, 10-bisphenylanthracene, coumarin, DAPI, dark quencher, DiOC6, dyghtuor, Fluo-3-fluoro, diaphora-3-fluorescein, Fluo-4, FluoProbes, fluorescein isothiocyanate, fluorescent image guided surgery, fluorojo-jade staining, fura-2-acetoxymethyl ester, GelGreen, GelRed, Green fluorescent protein, heptamethine (heptamethine) dye, India yellow, Indo-1, Fluorochrome yellow, fluorescein, MChery, Merocyanine, Nile blue, Nile Red, fluorescent whitening agent, perylene, phloxine, phycobilin, phycoerythrin, phycoerythrobilin, propidium iodide, fluorescent yellow dye (pyranine), rhodamine 123, rhodamine 6G, RiboGreen, RoGFP, Rose Benzorubine, rubrene, (E) -stilbene, (Z) -stilbene, sulforhodamine 101, sulforhodamine B, SYBR-I, synapto-butadiene, Tetraphenanthroline (sodium) bis (phenathionine) sulfonate (sodium salt of Tetraphenanthrene II), (E) -stilbene (sodium salt of Tetraphenanthroline (II), (Z) -bisodium sulfonate (sodium salt of sodium rhodine), Texas red, dadan yellow, TSQ, umbelliferone, yellow fluorescent protein, and YOYO-1.

Examples of photosensitive dyes for use according to the present disclosure include, but are not limited to: HpD, Porfimer sodium (Porfimer sodium) ((II))

Figure BDA0002311164830000061

Photosan) m-THPC, temoporfin

Figure BDA0002311164830000071

Verteporfin

Figure BDA0002311164830000072

HPPH

Figure BDA0002311164830000073

Palladium-bacteria-pheophorbide

Figure BDA0002311164830000074

5-ALA, 5-aminolevulinic acid

Figure BDA0002311164830000075

5-ALA methyl ester

Figure BDA0002311164830000076

5-ALA benzyl ester

Figure BDA0002311164830000077

5-ALA hexyl ester

Figure BDA0002311164830000078

Lutetium (III) -porphyrdelrin (texaphyrin) or Motexafin-lutetium (motexfin-lutetium)

Figure BDA0002311164830000079

SnET2, ethyl protoporphyrin tin (IV) (tin (IV) ethyl ethylpurpururin)

Figure BDA00023111648300000710

NPe6, mono-L-aspartic acid chloride e6, talaporfin sodium

Figure BDA00023111648300000711

BOPP, boronated protoporphyrinPhthalocyanine zinc

Figure BDA00023111648300000713

Phthalocyanine silicon

Figure BDA00023111648300000714

Sulfonated aluminium phthalocyanine derivative

Figure BDA00023111648300000715

ATMPn, acetoxy-tetrakis (. beta. -methoxyethyl-) porphinoid), TH9402 and dibromorhodamine methyl ester.

Examples of photoactive molecules for use in accordance with the present disclosure include those that are useful for fluorescence imaging (e.g., Near Infrared (NIR) fluorescent dyes), such as porphyrins, chlorophylls, chlorins, phthalocyanines, bacteriochlorins, deuteroporphyrins, porphyrins, and the like,(porfimer sodium),

Figure BDA00023111648300000717

(verteporfin),

Figure BDA00023111648300000718

/NPe6 (temoporfin), Foscan (talaporfin), methylene blue (Urolene)

Figure BDA00023111648300000719

Swiss blue, basic blue 9, Chromosmon, methylitonium chloride) and various derivatives of aminolevulinic acid (ALA, and/or ALA, and,

Figure BDA00023111648300000720

HexvixTM/

Figure BDA00023111648300000721

) Or a combination thereof. In some embodiments, the photosensitive molecule conjugated to the viral capsid protein assemblies of the present disclosure is a near-infrared dye. In some embodiments, the near infrared dye is

Figure BDA00023111648300000722

700DX。

According to aspects of the present disclosure, anti-cancer molecules (e.g., anti-cancer molecules) can be conjugated

Figure BDA00023111648300000723

700DX) to the capsid proteins of the viral capsid protein assemblies (e.g. papillomavirus L1 and/or L2 capsid proteins). In some embodiments, the anti-cancer molecule is covalently conjugated to capsid proteins of the viral capsid protein assembly. In some embodiments, the small anti-cancer molecule is covalently conjugated to a lysine residue of a capsid protein of a viral capsid protein assembly. Conjugation of anticancer molecules to capsid proteins does not impair the binding of the viral capsid protein assemblies to the surface of tumor cells, nor does it impair the binding of the viral capsid protein assemblies to HSPGs on the surface of tumor cells. The viral capsid protein assemblies conjugated to anti-cancer molecules may be referred to herein as "conjugates".

The papillomavirus L1 protein contains a large number of lysines that are available for chemical conjugation. Thus, a large number of photosensitive molecules (or other anti-cancer molecules) can be conjugated to one tumor-targeting viral capsid protein assembly of the present disclosure, i.e., the ratio of anti-cancer molecules to viral capsid protein assemblies can vary. In some embodiments, the ratio of viral capsid protein assemblies to photosensitive molecules is from about 1:10 to about 1:1000, or from about 1:50 to about 1: 1000. That is, in some embodiments, the viral capsid protein assembly may comprise from about 10 to about 1000 photosensitive molecules. In some embodiments, the ratio of viral capsid protein assemblies to photosensitive molecules is 1:10, 1:15, 1:20, 1:25, 1:50, 1:75, 1:100, 1:150, 1:200, 1:250, 1:500, 1:750, or 1: 1000. In some embodiments, the viral capsid protein assembly may comprise 10, 15, 20, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950 or 1000 photosensitive molecules. For example, a viral capsid protein assembly may comprise 10 to 1000, 10 to 500, 100 to 1000, 200 to 1000, 300 to 1000, 400 to 1000, 500 to 1000, 100 to 500, 200 to 500, 300 to 500, 400 to 500, 100 to 400, 200 to 400, 300 to 400, 100 to 300, or 200 to 300 photosensitive molecules. In some embodiments, the viral capsid protein assemblies may comprise more than 1000 photosensitive molecules or less than 10 anti-cancer molecules. Conjugation of photosensitive molecules to the L1 protein of the viral capsid protein assembly does not comprise the structural integrity or tumor targeting ability of the assembly, i.e. its ability to selectively bind HSPGs on the surface of tumor cells. Thus, another advantage of the tumor-targeting viral capsid protein assemblies of the present disclosure is their ability to specifically deliver large amounts of therapeutic agents to tumors even at very low molecular concentrations.

In some embodiments, the tumor-targeting viral capsid protein assembly comprises a photosensitive molecule conjugated to a viral capsid protein. In some embodiments, the photosensitive molecule is

Figure BDA0002311164830000081

700 DX. In some embodiments, the viral particle conjugate comprises an infrared-activated molecule (e.g., a peptide) and a pharmaceutically acceptable carrier700DX) such as a virus-like particle comprising 72 capsomers assembled from variant or modified HPV16/31L1 capsid protein and wild-type HPV L2 capsid protein. The conjugate has been shown to bind selectively to cancer cells and, after activation by a laser, selectively disrupt the membranes of cancer cells killing them without damaging adjacent normal cells.

The compositions of the present disclosure also comprise an immunotherapeutic agent for treating cancer. As used herein, "immunotherapeutic agent" refers to an agent that facilitates the use of the immune system to treat cancer. Such immunotherapeutics generally utilize the following facts: cancer cells typically have molecules on their surface that can be detected by the immune system. In some embodiments, the immunotherapeutic agent actively directs the immune system to attach tumor cells. In some embodiments, immunotherapeutics enhance existing anti-tumor responses, such as monoclonal antibodies, lymphocytes, and cytokines. In some embodiments, the immunotherapeutic agent modulates the activity of an immune checkpoint.

An "immune checkpoint" is a protein in the immune system that enhances or reduces an immune response signal (co-stimulatory molecule). Many cancers protect themselves from the immune system by inhibiting T cell signaling using inhibitory immune checkpoint proteins. Such inhibitory checkpoint proteins include, but are not limited to: cytotoxic T lymphocyte-associated protein 4(CTLA-4), programmed death 1 receptor (PD-1), T cell immunoglobulin and mucin domain 3(TIM3), lymphocyte activation gene 3(LAG3), T cell activation inhibitor 1 comprising a group V domain (VTVN1 or B7-H4), cluster of differentiation 276(CD276 or B7-H3), B and T Lymphocyte Attenuator (BTLA), galectin 9(GAL9), checkpoint kinase 1(Chk1), adenosine A2A receptor (A2aR), indoleamine 2, 3-bis (IDO), killer immunoglobulin-like receptor (KIR), lymphocyte activation gene 3(LAG3), and T cell activated group V domain Ig suppressor (VISTA).

Some of these immune checkpoint proteins require their respective binding partners or ligands for their immunosuppressive activity. For example, A2AR is a receptor for adenosine A2A, and binding of A2A to A2AR activates the negative immune feedback loop. As another example, PD-1 associates with its two ligands PD-L1 and PD-L2 to down-regulate the immune system by preventing T cell activation. PD-1 promotes programmed cell death of antigen-specific T cells in lymph nodes and simultaneously reduces programmed cell death of suppressor T cells, thereby fulfilling its immunosuppressive function. As another example, CTLA4 is present on the surface of T cells and, when bound to its binding partner CD80 or CD86 on the surface of Antigen Presenting Cells (APCs), transmits an inhibitory signal to the T cells, thereby reducing the immune response.

Cancer cells are known to use immune checkpoint proteins to evade attack by the immune system. Thus, the use of immune checkpoint inhibitors to enhance the immune response against cancer, and thus treat cancer, has been described. The immunotherapeutic agent in the compositions of the disclosure may also be an immune checkpoint inhibitor. In some embodiments, the immune checkpoint inhibits any one or more of: cytotoxic T lymphocyte-associated protein 4(CTLA-4), programmed death 1 receptor (PD-1), T cell immunoglobulin and mucin domain 3(TIM3), lymphocyte activation gene 3(LAG3), T cell activation inhibitor 1 comprising a group V domain (VTVN1 or B7-H4), cluster of differentiation 276(CD276 or B7-H3), B and T Lymphocyte Attenuator (BTLA), galectin 9(GAL9), checkpoint kinase 1(Chk1), adenosine A2A receptor (A2aR), indoleamine 2, 3-bis (IDO), killer immunoglobulin-like receptor (KIR), lymphocyte activation gene 3(LAG3), and T cell activated group V domain Ig suppressor (VISTA).

In some embodiments, "inhibit" means to prevent or attenuate binding of an immune checkpoint protein to its corresponding binding partner, e.g., PD-1, CTLA-4, or A2 aR. In some embodiments, the immune checkpoint inhibitor is an antibody. In some embodiments, the antibody comprises anti-CTLA-4, anti-PD-1, anti-PD-L1, anti-TIM 3, anti-LAG 3, anti-B7-H3, anti-B7-H4, anti-BTLA, anti-GAL 9, anti-Chk, anti-A2 aR, anti-IDO, anti-KIR, anti-LAG 3, anti-VISTA antibody, or a combination of any two or more of the foregoing antibodies. In some embodiments, the immunization isThe checkpoint inhibitor is a monoclonal antibody. In some embodiments, the immune checkpoint inhibitor comprises anti-PD 1, anti-PD-L1, anti-CTLA-4, or a combination of any two or more of the foregoing antibodies. For example, the anti-PD-1 antibody is pembrolizumab

Figure BDA0002311164830000101

Or nivolumab

Figure BDA0002311164830000102

And the anti-CTLA-4 antibody is ipilimumab

Figure BDA0002311164830000103

Thus, in some embodiments, the immune checkpoint inhibitor comprises pembrolizumab, nivolumab, ipilimumab, or any combination of two or more of the foregoing antibodies. It is to be understood that the examples described herein are not meant to be limiting, and that any immune checkpoint inhibitor known in the art, and any combination thereof, may be used in accordance with the present disclosure.

The compositions disclosed herein comprising tumor-targeting viral capsid protein assemblies and immunotherapeutic agents are useful for the treatment of cancer. Accordingly, the disclosure also encompasses methods of administering such compositions to a subject having a tumor to treat the tumor. The composition is administered to a subject in an amount effective to reduce tumor growth and/or prevent tumor recurrence.

By "administering" and variations thereof is meant providing a substance to a subject in a pharmacologically useful manner. The therapeutic agent or composition may be administered to the subject using conventional methods known to those of ordinary skill in the medical arts, depending on the type of cancer to be treated or the site of the cancer. The composition may also be administered by other conventional routes, such as orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally, or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intraarterial, intravesical, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques. In addition, they can be administered to a subject by injectable depot routes of administration, e.g., using 1 month, 3 months, or 6 months depot injectable or biodegradable materials and methods. In some embodiments, the composition can be administered systemically, e.g., intravenously. In some embodiments, the composition is administered topically. In some embodiments, the composition is administered by implantation.

In some embodiments, the compositions of the present disclosure can be injected into a target site where tumor growth is present. Injection methods that may be used in accordance with the present disclosure include, but are not limited to, intravenous injection, intralesional injection, subcutaneous injection, intravitreal injection, suprachoroidal injection, intraperitoneal injection, intraarterial injection, intrahepatic injection, and intravesical injection. In the case of using an injection, a hollow needle, a coated needle, a small needle, or a microneedle is used according to the injection area.

In some embodiments, the methods of cancer treatment of the present disclosure comprise administering to a subject having a tumor a composition comprising a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein, and a composition comprising an immunotherapeutic agent.

In some embodiments, the tumor-targeting viral capsid protein assemblies and the immunotherapeutic agent are administered simultaneously. By "simultaneously" is meant that two or more materials/agents (e.g., tumor-targeting viral capsid protein assemblies and immunotherapeutic agents) are administered to a subject simultaneously. For example, compositions comprising tumor-targeting viral capsid protein assemblies or immunotherapeutic agents can be combined/mixed prior to administration.

In some embodiments, the tumor-targeting viral capsid protein assemblies and the immunotherapeutic agent are administered sequentially. By "sequentially" is meant that administration of one agent and administration of another agent are separated in time (in two separate steps). In some embodiments, the tumor-targeting viral capsid protein assemblies are administered first, and then the immunotherapeutic agent (e.g., checkpoint inhibitor) is administered. In some embodiments, the immunotherapeutic agent is administered first, and then the tumor-targeting viral capsid protein assemblies are administered. The time between two steps of the administering step may be at least 1 minute, at least 5 minutes, at least 30 minutes, at least 1 hour, at least 5 hours, at least 10 hours, at least 1 day, at least 1 week, or even at least one month. When the tumor-targeting viral capsid protein assemblies and immunotherapeutic agent are administered sequentially, they can also be administered by different routes or at different locations. For example, tumor-targeting viral capsid protein assemblies can be administered intralesionally to exposed tumor lesions or subcutaneously to the skin of a subject, while immunotherapeutics can be administered systemically, e.g., intravenously.

In some embodiments, the cancer treatment methods of the present disclosure comprise administering an immunotherapeutic agent to a subject having a tumor undergoing treatment with a tumor-targeting viral capsid protein assembly comprising a molecule (e.g., a photosensitive molecule) conjugated to a viral capsid protein.

Depending on the type of molecule, the photoactive molecules of the present disclosure may be activated by infrared, near infrared, or ultraviolet light. For example, in some embodiments, infrared, near-infrared, or ultraviolet lasers can be used to activate photosensitive molecules of tumor-targeted viral capsid protein assemblies. The energy delivered by the laser may be 5 joules (J) to about 150J, or 8J to 36J. In some embodiments, the energy delivered by the laser is 5J, 6J, 7J, 8J, 9J, 10J, 12J, 14J, 16J, 18J, 20J, 22J, 24J, 26J, 28J, 30J, 32J, 34J, 36J, 38J, 40J, 50J, 60J, 70J, 80J, 90J, 100J, 110J, 120J, 130J, 140J, or 150J.

Light or laser light may be applied to the photosensitive molecules (or tumor-targeting viral capsid protein assemblies) for about 5 seconds to about 5 minutes. For example, in some embodiments, light or laser is applied to the photosensitive molecules for 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or 55 seconds to activate the molecules. In some embodiments, the laser is applied to the photosensitive molecules for 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, or 5 minutes.

Light or laser can be applied to the photosensitive molecule (or tumor-targeted viral capsid protein assembly) from about 30 minutes to about 48 hours after administration of the tumor-targeted viral capsid protein assembly. For example, in some embodiments, light or laser is applied to the photosensitive molecule 30, 35, 40, 45, 50, or 55 minutes after administration of the tumor-targeting viral capsid protein assembly. In some embodiments, the light or laser is applied to the photosensitive molecule 1, 2,3, 4, 5, 6, 7, 8, 9,10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours after administration of the tumor-targeting viral capsid protein assembly. In some embodiments, light or laser is applied to the photosensitive molecule 36 or 48 hours after administration of the tumor-targeting viral capsid protein assemblies.

For example, light or laser light may be applied directly to the tumor site.

In accordance with the cancer treatment methods of the present disclosure, immunotherapeutic agents, such as immune checkpoint inhibitors, may be further administered to a subject suffering from a tumor undergoing treatment with a viral capsid protein assembly conjugated to a photosensitive molecule. Any of the immunotherapeutic agents described herein are suitable for administration to a subject. It will be appreciated that different immunotherapeutic agents may be administered depending on the type of tumor to be treated. One skilled in the art will be able to determine the appropriate immunotherapeutic agent. In some embodiments, an immune checkpoint inhibitor, e.g., an anti-PD-1, anti-PD-L1, and/or anti-CTLA-4 antibody, is administered.

Tumor types that can be treated using the compositions and methods of the present disclosure include, but are not limited to, premalignant tumors, malignant tumors, metastases, or any disease or condition characterized by uncontrolled cell growth such that it is considered cancerous or precancerous. The cancer may be primary or metastatic cancer. Cancers include, but are not limited to: eye cancer, biliary tract cancer, bladder cancer, pleural cancer, gastric cancer, ovarian cancer, meningeal cancer, kidney cancer, brain cancer including glioblastoma and medulloblastoma, breast cancer, cervical cancer, choriocarcinoma, colon cancer, endometrial cancer, esophageal cancer, stomach cancer, tumors of the blood system including acute lymphocytic and myelogenous leukemias (hematogenous neoplasms), multiple myeloma, AIDS-related leukemia and adult T-cell leukemia lymphoma, intraepithelial tumors including Bowen's disease and Paget's disease, liver cancer, lung cancer, lymphoma including Hodgkin's disease and lymphocytic lymphoma, neuroblastoma, oral cancer including squamous cell carcinoma, ovarian cancer including those produced by epithelial cells, stromal cells, germ cells and mesenchymal cells, pancreatic cancer, prostate cancer, rectal cancer, sarcomas including leiomyosarcoma, rhabdomyosarcoma, liposarcoma, fibrosarcoma, and osteosarcoma, skin cancers including melanoma, kaposi's sarcoma, basal cell carcinoma, and squamous cell carcinoma, testicular cancers including germ tumors (germinal tumors) such as seminoma, non-seminoma, teratoma, choriocarcinoma, interstitial tumors, and germ cell tumors, thyroid cancers including thyroid adenocarcinoma and medullary carcinoma, and renal cancers including adenocarcinoma and Wilm's tumor. Common cancers include breast, prostate, lung, ovarian, colorectal, and brain cancers. In some embodiments, the tumor is melanoma, carcinoma, sarcoma, or lymphoma.

In some embodiments, the tumor has metastasized and it has a skin or surface exposed tumor lesion (e.g., Merkel cell carcinoma, head and neck squamous cell carcinoma, basal cell carcinoma, breast and metastatic breast cancers, cutaneous T-cell lymphoma (Sezary syndrome), and sarcoma). In some embodiments, a tumor, such as a tumor located in the head, neck, cervix, larynx, esophagus, or skin, is accessible without surgical intervention. In some embodiments, the tumor is cancerous or malignant. In some embodiments, the tumor is accessible (light/laser accessible) by surgical or endoscopic intervention. In some embodiments, the tumor is metastatic. In some embodiments, the tumor is a primary tumor and has metastasized. In some embodiments, the metastatic cancer has a tumor lesion that is exposed to the skin or surface. In some embodiments, the tumor has a lesion that can be treated by an infrared laser. In some embodiments, the metastatic cancer has a tumor focus or metastasis in the eye. In some embodiments, the tumor is an eye tumor and has metastasized to the liver. When the tumor is or has metastasized to the eye, the ocular tumor is located in the vitreous, suprachoroidal space, iris, ciliary body, sclera, fovea, retina, optic disc, or optic nerve.

In some embodiments, the subject having a tumor is a mammal, e.g., a human.

The compositions and/or therapeutic agents of the present disclosure are administered to a subject in an effective amount to reduce or prevent the growth of cancer. As used herein, "effective amount" refers to the amount of each agent required to confer a therapeutic effect on a subject, either alone or in combination with one or more other agents. As will be recognized by those of skill in the art, an effective amount will depend upon the particular condition being treated, the severity of the condition, the individual subject parameters (including age, physical condition, size, sex and weight, duration of treatment, nature of concurrent therapy (if any), specific route of administration, and the like, and factors similar to those known to and skilled in the art.

Empirical considerations such as half-life will often aid in determining the dosage. For example, agents compatible with the human immune system, such as agents comprising regions from humanized antibodies or fully human antibodies, can be used to extend the half-life of the compound and prevent the compound from being attacked by the host's immune system. The frequency of administration can be determined and adjusted during the course of treatment, and is typically, but not necessarily, based on the treatment and/or inhibition and/or amelioration and/or delay of the disease. Alternatively, sustained continuous release formulations of the compounds may be suitable. Various formulations and devices for achieving sustained release are known in the art.

In some embodiments, the dose is once daily, every other day, every third day, every fourth day, every fifth day, or every sixth day. In some embodiments, the frequency of administration is once per week, once every 2 weeks, once every 4 weeks, once every 5 weeks, once every 6 weeks, once every 7 weeks, once every 8 weeks, once every 9 weeks, or once every 10 weeks; or once a month, once every 2 months, or once every 3 months or longer. The course of such treatment is readily monitored by conventional techniques and assays. The dosage regimen, including the compound used, may vary over time.

In some embodiments, for adult subjects of normal weight, a dose of about 0.01 to 1000mg/kg may be administered. In some embodiments, the dose is 1 to 200 mg. The particular dosage regimen, i.e., dosage, timing and repetition, will depend on the particular subject and the subject's medical history, as well as the nature of the agent (e.g., the half-life of the agent and other art-recognized factors).

For the purposes of this disclosure, the appropriate dosage of a therapeutic agent as described herein will depend on the particular agent (or composition thereof) used, the formulation and route of administration, the type and severity of the disease, whether the compound is being used for prophylactic or therapeutic purposes, previous therapy, the subject's clinical history and response to the antagonist, and the discretion of the attending physician. Typically, the clinician will administer the agent until a dosage is reached that achieves the desired result. Administration of one or more agents may be continuous or intermittent, depending on, for example, the physiological condition of the recipient, whether the purpose of administration is therapeutic or prophylactic, and other factors known to the skilled artisan. Administration of the agent may be substantially continuous over a preselected period of time, or may be a series of spaced doses, for example before, during or after the onset of disease.

The term "treatment" as used herein refers to the application or administration of an agent or a composition comprising an agent to a subject suffering from or having a disease, a symptom of a disease, or a predisposition toward a disease, for the purpose of treating, curing, alleviating, altering, remedying, ameliorating, improving, or affecting the disease, the symptom of the disease, or the predisposition toward the disease.

Alleviating a disease includes delaying the development or progression of the disease, or reducing the severity of the disease. Relief from the disease does not necessarily require a therapeutic outcome. As used herein, "delaying the progression of" a disease means delaying, impeding, slowing, delaying, stabilizing and/or delaying the progression of the disease. The length of time for such a delay may vary depending on the history of the disease and/or the individual undergoing treatment. A method of "delaying" or reducing the progression of a disease or delaying the onset of a disease is a method that reduces the likelihood of developing one or more symptoms of a disease within a given time frame and/or reduces the extent of symptoms within a given time frame compared to not using the method. Such comparisons are typically based on clinical studies, using a number of subjects sufficient to give statistically significant results.

"progression" or "progression" of a disease means the initial manifestation and/or subsequent progression of the disease. Development of the disease can be detected and assessed using standard clinical techniques well known in the art. However, occurrence also refers to progression that may not be detectable. For the purposes of this disclosure, onset or progression refers to the biological process of a symptom. "development" includes occurrence, recurrence and onset. As used herein, "onset" or "occurrence" of a disease includes initial onset and/or recurrence.

The present disclosure also encompasses the following numbered paragraphs:

1. a process which comprises

(a) Administering to a subject having a tumor a composition comprising a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of said assembly; and

(b) administering a composition comprising an immune checkpoint inhibitor to a subject having a tumor.

2. The method of paragraph 1, wherein the viral capsid protein is a human papilloma virus capsid protein.

3. The method of paragraph 1, wherein the viral capsid protein is a non-human papilloma virus capsid protein.

4. The method of paragraph 3, wherein the non-human papillomavirus capsid protein is a bovine papillomavirus capsid protein or a cotton-tail rabbit papillomavirus protein.

5. The method of any of paragraphs 1 to 4, wherein the viral capsid protein comprises a papillomavirus L1 capsid protein or a combination of a papillomavirus L1 capsid protein and a papillomavirus L2 capsid protein.

6. The method of any of paragraphs 1 to 5, wherein the tumor-targeting viral capsid protein assemblies are capsomeres or virus-like particles.

7. The method of any of paragraphs 1 to 6, wherein the tumor-targeting viral capsid protein assembly comprises from 10 to 1000 photosensitive molecules, optionally wherein the tumor-targeting viral capsid protein assembly comprises from 200 to 500 photosensitive molecules.

8. The method of any of paragraphs 1 to 7, wherein the photosensitive molecule comprises a fluorescent dye, an Infrared (IR) dye, a Near Infrared (NIR) dye, a porphyrin, a chlorophyll, a chlorin, a phthalocyanine, a bacteriochlorin, a deuteroporphyrin, a porfimer sodium, verteporfin, temoporfin, talaporfin, methylene blue, aminolevulinic acid, or a combination thereof.

9. The method of any of paragraphs 1 to 8, wherein the tumor-targeting viral capsid protein assembly is a virus-like particle comprising 72 capsomers assembled from variant or modified HPV16/31L1 capsid protein and wild-type HPV L2 capsid protein, and wherein the photosensitive molecule comprises IRdye700 DX.

10. The method of any of paragraphs 1 to 9, wherein the immune checkpoint inhibitor inhibits binding of CTLA-4, PD-1, PD-L1, TIM3, LAG3, B7-H3, B7-H4, BTLA, GAL9, Chk1, or A2aR to the respective binding partner.

11. The method of paragraph 10, wherein the immune checkpoint inhibitor is an antibody.

12. The method of paragraph 11 wherein the antibody is a monoclonal antibody.

13. The method of any one of paragraphs 1 to 12, wherein the antibody is selected from the group consisting of an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM 3 antibody, an anti-LAG 3 antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-BTLA antibody, an anti-GAL 9 antibody, an anti-Chk 1 antibody, an anti-A2 aR antibody, and a combination of any two or more of the foregoing antibodies.

14. The method of paragraph 13, wherein the antibody is selected from pembrolizumab, nivolumab, ipilimumab, and any combination of two or more of the foregoing antibodies.

15. The method of any of paragraphs 1 to 14, wherein the tumor-targeting viral capsid protein assemblies and the immune checkpoint inhibitor are administered simultaneously.

16. The method of any of paragraphs 1 to 14, wherein the tumor-targeting viral capsid protein assemblies and the immune checkpoint inhibitor are administered sequentially.

17. The method of paragraph 16, wherein the tumor-targeting viral capsid protein assemblies are administered to the subject prior to administration of the immune checkpoint inhibitor to the subject.

18. The method of paragraphs 16 and 17, wherein the tumor-targeting viral capsid protein assemblies are administered locally to the tumor or tumor lesion, and wherein the immune checkpoint inhibitor is administered systemically.

19. The method of any of paragraphs 1 to 18, wherein the tumor-targeting viral capsid protein assemblies and/or immune checkpoint inhibitor are administered by injection, topically or by implantation.

20. The method of paragraph 19, wherein the injection is intralesional injection, subcutaneous injection, intravitreal injection, suprachoroidal injection, intraperitoneal injection, intraarterial injection, intrahepatic injection, intravesical injection, or any combination thereof.

21. The method of any of paragraphs 1 to 20, further comprising activating the photosensitive molecules using an infrared laser, a near-infrared laser, or an ultraviolet laser.

22. The method of any of paragraphs 1 to 21, wherein the tumor is cancerous.

23. The method of paragraph 23, wherein the tumor is metastatic.

24. The method of any of paragraphs 1 to 23, wherein the tumor is melanoma, carcinoma, sarcoma, or lymphoma.

25. The method of any of paragraphs 1 to 24, wherein the tumor has a lesion treatable with an infrared laser.

26. The method of paragraph 25, wherein the tumor is merkel cell carcinoma, squamous cell carcinoma, basal cell carcinoma, metastatic breast cancer, cutaneous T-cell lymphoma or sarcoma.

27. The method of any of paragraphs 1 to 25, wherein the tumor is a primary tumor and has metastasized.

28. The method of any of paragraphs 1 to 27, wherein the tumor is located and/or has metastasized to: liver, bladder, eye, head, neck, cervix, larynx, skin, lung, pleura, pancreas, stomach, esophagus, colon, breast, ovary, prostate, testis, brain, meninges, kidney.

29. The method of any one of paragraphs 1 to 27, wherein the subject is a human.

30. A method comprising administering an immune checkpoint inhibitor to a subject having a tumor undergoing treatment with a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of said assembly.

31. A composition comprising an immune checkpoint inhibitor and a tumor-targeting viral capsid protein assembly comprising a photosensitive molecule conjugated to a viral capsid protein of said assembly.

32. The composition of paragraph 31, wherein the viral capsid protein is a human papilloma virus capsid protein. 33. The composition of paragraph 31, wherein the viral capsid protein is a non-human papilloma virus capsid protein.

34. The composition of paragraph 33, wherein the non-human papillomavirus capsid protein is a bovine papillomavirus capsid protein or a cotton-tail rabbit papillomavirus protein.

35. The composition of any of paragraphs 31 to 34, wherein the viral capsid protein comprises a papillomavirus L1 capsid protein or a combination of a papillomavirus L1 capsid protein and a papillomavirus L2 capsid protein.

36. The composition of any of paragraphs 31 to 35, wherein the tumor-targeting viral capsid protein assemblies are capsomeres or virus-like particles.

37. The composition of any of paragraphs 31 to 36, wherein the tumor-targeting viral capsid protein assembly comprises 10 to 1000 photosensitive molecules.

38. The composition of any of paragraphs 31 to 37, wherein the photosensitive molecule comprises a fluorescent dye, an Infrared (IR) dye, a Near Infrared (NIR) dye, a porphyrin, a chlorophyll, a chlorin, a phthalocyanine, a bacteriochlorin, a deuteroporphyrin, a porfimer sodium, a verteporfin, a temoporfin, a talaporfin, a methylene blue, an aminolevulinic acid, or a combination thereof.

39. The composition of any of paragraphs 31 to 38, wherein the tumor-targeting viral capsid protein assembly is a virus-like particle comprising 72 capsomers assembled from variant or modified HPV16/31L1 capsid protein and wild-type HPV L2 capsid protein, and wherein the photosensitive molecule comprises IRdye700 DX.

40. The composition of any one of paragraphs 31 to 39, wherein the immune checkpoint inhibitor inhibits binding of CTLA-4, PD-1, PD-L1, TIM3, LAG3, B7-H3, B7-H4, BTLA, GAL9, Chk1, or A2aR to a respective binding partner.

41. The composition of paragraph 40, wherein the immune checkpoint inhibitor is an antibody.

42. The composition of paragraph 41 wherein the antibody is a monoclonal antibody.

43. The composition of any one of paragraphs 31 to 42, wherein the antibody is selected from the group consisting of an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM 3 antibody, an anti-LAG 3 antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-BTLA antibody, an anti-GAL 9 antibody, an anti-Chk 1 antibody, an anti-A2 aR antibody, and a combination of any two or more of the foregoing antibodies.

44. The composition of paragraph 43, wherein the antibody is selected from pembrolizumab, nivolumab, ipilimumab, and any combination of two or more of the foregoing antibodies.

The disclosure is further illustrated by the following examples, which are in no way to be construed as further limiting.

Examples

24页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:高折射率、高阿贝数的人工晶状体材料

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!