JPWO2021138264A5 - - Google Patents

Download PDF

Info

Publication number
JPWO2021138264A5
JPWO2021138264A5 JP2022539657A JP2022539657A JPWO2021138264A5 JP WO2021138264 A5 JPWO2021138264 A5 JP WO2021138264A5 JP 2022539657 A JP2022539657 A JP 2022539657A JP 2022539657 A JP2022539657 A JP 2022539657A JP WO2021138264 A5 JPWO2021138264 A5 JP WO2021138264A5
Authority
JP
Japan
Prior art keywords
antibody
composition
seq
chain variable
variable region
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
JP2022539657A
Other languages
Japanese (ja)
Other versions
JP2023508496A (en
Publication date
Application filed filed Critical
Priority claimed from PCT/US2020/067173 external-priority patent/WO2021138264A1/en
Publication of JP2023508496A publication Critical patent/JP2023508496A/en
Publication of JPWO2021138264A5 publication Critical patent/JPWO2021138264A5/ja
Pending legal-status Critical Current

Links

Description

本発明は、以下の実施例を参照することによってより十分に理解されるであろう。しかし、それらは本発明の範囲の制限と解釈されてはならない。本明細書で記載される実施例及び実施形態は、単に例示目的であり、それを考慮して種々の改変又は変更が当業者に示唆され、本出願及び添付の特許請求の範囲の趣旨及び範囲内に含まれるべきであるということは理解される。
例えば、本発明は以下の実施形態を包含する:
[実施形態1]対象においてCD70発現がんを処置する方法であって、対象に非フコシル化抗CD70抗体の治療有効量を投与することを含み、前記方法は、対象においてがん細胞の枯渇をもたらし、対象においてCD70+制御性T細胞(CD70+Treg)の枯渇をもたらさず、抗CD70抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含み、がんが骨髄異形成症候群(MDS)及び急性骨髄性白血病(AML)からなる群から選択される、方法。
[実施形態2]抗CD70抗体が、配列番号1のアミノ酸配列に対して少なくとも85%同一のアミノ酸配列を含む重鎖可変領域及び配列番号2のアミノ酸配列に対して少なくとも85%同一のアミノ酸配列を含む軽鎖可変領域を含む、実施形態1に記載の方法。
[実施形態3]抗CD70抗体が、配列番号1のアミノ酸配列を含む重鎖可変領域及び配列番号2のアミノ酸配列を含む軽鎖可変領域を含む、実施形態1に記載の方法。
[実施形態4]Fcドメインが、抗体依存性細胞傷害(ADCC)、抗体依存性細胞貪食(ADCP)及び補体依存性細胞傷害(CDC)のうち1つ以上を媒介する抗体エフェクタードメインである、実施形態1から3のいずれかに記載の方法。
[実施形態5]Fcドメインが、ADCCを媒介する抗体エフェクタードメインである、実施形態1から3のいずれかに記載の方法。
[実施形態6]FcドメインがヒトFcドメインである、実施形態1から5のいずれかに記載の方法。
[実施形態7]抗CD70抗体がボルセツズマブである、実施形態1から6のいずれかに記載の方法。
[実施形態8]抗体が治療剤にコンジュゲートされている、実施形態1から7のいずれかに記載の方法。
[実施形態9]治療剤が、化学療法剤又は免疫調節剤である、実施形態8に記載の方法。
[実施形態10]治療剤が化学療法剤である、実施形態8に記載の方法。
[実施形態11]化学療法剤が、モノメチルアウリスタチンE(MMAE)又はモノメチルアウリスタチンF(MMAF)である、実施形態10に記載の方法。
[実施形態12]治療剤が免疫調節剤である、実施形態8に記載の方法。
[実施形態13]前記方法が抗CD70抗体の集団を投与することを含み、抗CD70抗体の集団中の各抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含み、抗CD70抗体の集団中の抗CD70抗体の少なくとも50%がコアフコシル化を欠く、実施形態1から12のいずれかに記載の方法。
[実施形態14]抗CD70抗体の集団中の抗CD70抗体の少なくとも70%が、コアフコシル化を欠く、実施形態13に記載の方法。
[実施形態15]抗CD70抗体の集団中の抗CD70抗体の少なくとも90%が、コアフコシル化を欠く、実施形態13に記載の方法。
[実施形態16]がんがMDSである、実施形態1から15のいずれかに記載の方法。
[実施形態17]MDSが再発性又は難治性MDSである、実施形態16に記載の方法。
[実施形態18]対象が、MDSのための以前の低メチル化剤(HMA)療法後に処置失敗を経験した、実施形態17に記載の方法。
[実施形態19]がんがAMLである、実施形態1から15のいずれかに記載の方法。
[実施形態20]AMLが再発性又は難治性AMLである、実施形態19に記載の方法。
[実施形態21]対象が、AMLを処置するために2つの以前の処置レジメンを受けた、実施形態20に記載の方法。
[実施形態22]対象が、AMLを処置するために3つの以前の処置レジメンを受けた、実施形態20に記載の方法。
[実施形態23]がん細胞の少なくとも約0.1%、少なくとも約1%、少なくとも約2%、少なくとも約3%、少なくとも約4%、少なくとも約5%、少なくとも約6%、少なくとも約7%、少なくとも約8%、少なくとも約9%、少なくとも約10%、少なくとも約15%、少なくとも約20%、少なくとも約25%、少なくとも約30%、少なくとも約35%、少なくとも約40%、少なくとも約45%、少なくとも約50%、少なくとも約60%、少なくとも約70%又は少なくとも約80%が、CD70を発現する、実施形態1から22のいずれかに記載の方法。
[実施形態24]対象に非フコシル化抗CD70抗体を投与することが、対象に非フコシル化抗CD70抗体を投与する前のがん細胞の量と比較して、少なくとも約5%、少なくとも約6%、少なくとも約7%、少なくとも約8%、少なくとも約9%、少なくとも約10%、少なくとも約15%、少なくとも約20%、少なくとも約25%、少なくとも約30%、少なくとも約35%、少なくとも約40%、少なくとも約45%、少なくとも約50%、少なくとも約60%、少なくとも約70%、少なくとも約80%、少なくとも約90%、少なくとも約95%又は約100%のがん細胞の枯渇をもたらす、実施形態1から23のいずれかに記載の方法。
[実施形態25]対象に非フコシル化抗CD70抗体を投与することが、対象に脱フコシル化抗CD70抗体を投与する前のCD70+Tregの量と比較して、約20%、約10%、約9%、約8%、約7%、約6%、約5%、約4%、約3%、約2%、約1%又は約0.1%以下のCD70+Tregの枯渇をもたらす、実施形態1から24のいずれかに記載の方法。
[実施形態26]ベースラインに対して、非フコシル化抗CD70抗体の投与後に対象において1つ以上の治療効果が改善される、実施形態1から25のいずれかに記載の方法。
[実施形態27]1つ以上の治療効果が、客観的奏効率、応答の持続期間、応答までの期間、無増悪生存期間及び全生存期間からなる群から選択される、実施形態26に記載の方法。
[実施形態28]客観的奏効率が、少なくとも約20%、少なくとも約25%、少なくとも約30%、少なくとも約35%、少なくとも約40%、少なくとも約45%、少なくとも約50%、少なくとも約60%、少なくとも約70%又は少なくとも約80%である、実施形態1から27のいずれかに記載の方法。
[実施形態29]対象が、非フコシル化抗CD70抗体の投与後に少なくとも約1カ月、少なくとも約2カ月、少なくとも約3カ月、少なくとも約4カ月、少なくとも約5カ月、少なくとも約6カ月、少なくとも約7カ月、少なくとも約8カ月、少なくとも約9カ月、少なくとも約10カ月、少なくとも約11カ月、少なくとも約12カ月、少なくとも約18カ月、少なくとも約2年、少なくとも約3年、少なくとも約4年又は少なくとも約5年の無増悪生存期間を示す、実施形態1から28のいずれかに記載の方法。
[実施形態30]対象が、非フコシル化抗CD70抗体の投与後に少なくとも約1カ月、少なくとも約2カ月、少なくとも約3カ月、少なくとも約4カ月、少なくとも約5カ月、少なくとも約6カ月、少なくとも約7カ月、少なくとも約8カ月、少なくとも約9カ月、少なくとも約10カ月、少なくとも約11カ月、少なくとも約12カ月、少なくとも約18カ月、少なくとも約2年、少なくとも約3年、少なくとも約4年又は少なくとも約5年の全生存期間を示す、実施形態1から29のいずれかに記載の方法。
[実施形態31]抗CD70抗体に対する応答の持続期間が、非フコシル化抗CD70抗体の投与後に少なくとも約1カ月、少なくとも約2カ月、少なくとも約3カ月、少なくとも約4カ月、少なくとも約5カ月、少なくとも約6カ月、少なくとも約7カ月、少なくとも約8カ月、少なくとも約9カ月、少なくとも約10カ月、少なくとも約11カ月、少なくとも約12カ月、少なくとも約18カ月、少なくとも約2年、少なくとも約3年、少なくとも約4年又は少なくとも約5年である、実施形態1から30のいずれかに記載の方法。
[実施形態32]抗CD70抗体の投与経路が静脈内である、実施形態1から31のいずれかに記載の方法。
[実施形態33]対象がヒトである、実施形態1から32のいずれかに記載の方法。
[実施形態34]抗CD70抗体が、アザシチジンと組み合わせて投与される、実施形態1から33のいずれかに記載の方法。
[実施形態35]抗CD70抗体が、ベネトクラクスと組み合わせて投与される、実施形態1から33のいずれかに記載の方法。
[実施形態36]抗CD70抗体が、アザシチジン及びベネトクラクスと組み合わせて投与される、実施形態1から33のいずれかに記載の方法。
[実施形態37]抗CD70抗体が、フルオロキノロン(fluoroquinalone)と組み合わせて投与される、実施形態1から35のいずれかに記載の方法。
[実施形態38]CD70発現がんの処置のための医薬組成物であって、非フコシル化抗CD70抗体と、少なくとも1つの薬学的に適合する成分とを含み、抗CD70抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含み、前記組成物が、実施形態1から37のいずれかに記載の方法における使用のためのものである、医薬組成物。
[実施形態39]非フコシル化抗CD70抗体と、実施形態1から37のいずれかに記載の方法において抗CD70抗体を使用するための使用説明書とを含むキットであって、抗CD70抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含む、キット。
The invention will be more fully understood by reference to the following examples. However, they should not be construed as limiting the scope of the invention. The examples and embodiments described herein are for illustrative purposes only, and various modifications or changes may be suggested to those skilled in the art in light of the same and within the spirit and scope of this application and the appended claims. It is understood that it should be included within.
For example, the invention encompasses the following embodiments:
[Embodiment 1] A method of treating a CD70-expressing cancer in a subject, the method comprising administering to the subject a therapeutically effective amount of a non-fucosylated anti-CD70 antibody, said method causing depletion of cancer cells in the subject. and does not result in depletion of CD70+ regulatory T cells (CD70+ Tregs) in the subject, and the anti-CD70 antibody is a heavy antibody containing three CDRs of SEQ ID NO: 1, where the CDRs of the anti-CD70 antibody are defined by the Kabat numbering scheme. a chain variable region, a light chain variable region comprising the three CDRs of SEQ ID NO: 2, and an Fc domain, wherein the cancer is selected from the group consisting of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). .
[Embodiment 2] An anti-CD70 antibody has a heavy chain variable region comprising an amino acid sequence that is at least 85% identical to the amino acid sequence of SEQ ID NO: 1 and an amino acid sequence that is at least 85% identical to the amino acid sequence of SEQ ID NO: 2. 2. The method of embodiment 1, comprising a light chain variable region comprising:
[Embodiment 3] The method according to Embodiment 1, wherein the anti-CD70 antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 1 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 2.
[Embodiment 4] The Fc domain is an antibody effector domain that mediates one or more of antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cellular cytotoxicity (CDC), The method according to any of embodiments 1-3.
[Embodiment 5] The method according to any one of Embodiments 1 to 3, wherein the Fc domain is an antibody effector domain that mediates ADCC.
[Embodiment 6] The method according to any one of Embodiments 1 to 5, wherein the Fc domain is a human Fc domain.
[Embodiment 7] The method according to any one of Embodiments 1 to 6, wherein the anti-CD70 antibody is borcetuzumab.
[Embodiment 8] The method of any of embodiments 1 to 7, wherein the antibody is conjugated to a therapeutic agent.
[Embodiment 9] The method according to Embodiment 8, wherein the therapeutic agent is a chemotherapeutic agent or an immunomodulator.
[Embodiment 10] The method according to Embodiment 8, wherein the therapeutic agent is a chemotherapeutic agent.
[Embodiment 11] The method according to Embodiment 10, wherein the chemotherapeutic agent is monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF).
[Embodiment 12] The method of embodiment 8, wherein the therapeutic agent is an immunomodulator.
[Embodiment 13] The method comprises administering a population of anti-CD70 antibodies, each antibody in the population of anti-CD70 antibodies having a CDR of SEQ ID NO: 1, wherein the CDRs of the anti-CD70 antibody are defined by the Kabat numbering scheme. A heavy chain variable region comprising three CDRs, a light chain variable region comprising three CDRs of SEQ ID NO: 2, and an Fc domain, and at least 50% of the anti-CD70 antibodies in the population of anti-CD70 antibodies lack core fucosylation. The method according to any of Forms 1 to 12.
[Embodiment 14] The method of embodiment 13, wherein at least 70% of the anti-CD70 antibodies in the population of anti-CD70 antibodies lack core fucosylation.
[Embodiment 15] The method of embodiment 13, wherein at least 90% of the anti-CD70 antibodies in the population of anti-CD70 antibodies lack core fucosylation.
[Embodiment 16] The method according to any one of Embodiments 1 to 15, wherein the cancer is MDS.
[Embodiment 17] The method according to Embodiment 16, wherein the MDS is relapsed or refractory MDS.
[Embodiment 18] The method of embodiment 17, wherein the subject experienced treatment failure after previous hypomethylating agent (HMA) therapy for MDS.
[Embodiment 19] The method according to any one of Embodiments 1 to 15, wherein the cancer is AML.
[Embodiment 20] The method according to Embodiment 19, wherein the AML is relapsed or refractory AML.
[Embodiment 21] The method of embodiment 20, wherein the subject has received two previous treatment regimens to treat AML.
[Embodiment 22] The method of embodiment 20, wherein the subject has received three previous treatment regimens to treat AML.
[Embodiment 23] At least about 0.1%, at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least 23. The method of any of embodiments 1-22, wherein about 50%, at least about 60%, at least about 70% or at least about 80% express CD70.
[Embodiment 24] Administering the non-fucosylated anti-CD70 antibody to the subject results in a reduction in the amount of cancer cells of at least about 5%, at least about 6% compared to the amount of cancer cells before administering the non-fucosylated anti-CD70 antibody to the subject. %, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40 %, at least about 45%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95% or about 100% depletion of cancer cells. The method according to any of Forms 1 to 23.
[Embodiment 25] Administering the non-fucosylated anti-CD70 antibody to the subject reduces the amount of CD70+ Tregs by about 20%, about 10%, compared to the amount of CD70+ Tregs before administering the defucosylated anti-CD70 antibody to the subject. Implementation that results in depletion of CD70+ Tregs of less than about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1%, or about 0.1% The method according to any of Forms 1 to 24.
[Embodiment 26] The method of any of embodiments 1-25, wherein one or more therapeutic effects are improved in the subject after administration of the non-fucosylated anti-CD70 antibody relative to baseline.
[Embodiment 27] The method according to embodiment 26, wherein the one or more therapeutic effects are selected from the group consisting of objective response rate, duration of response, time to response, progression-free survival, and overall survival. Method.
[Embodiment 28] The objective response rate is at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%. , at least about 70% or at least about 80%.
[Embodiment 29] The subject is at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months after administration of the non-fucosylated anti-CD70 antibody. at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about 12 months, at least about 18 months, at least about 2 years, at least about 3 years, at least about 4 years, or at least about 5 29. The method of any of embodiments 1-28, showing progression free survival in years.
[Embodiment 30] The subject is at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months after administration of the non-fucosylated anti-CD70 antibody. at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about 12 months, at least about 18 months, at least about 2 years, at least about 3 years, at least about 4 years, or at least about 5 30. The method of any of embodiments 1-29, showing overall survival in years.
[Embodiment 31] The duration of the response to the anti-CD70 antibody is at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about 12 months, at least about 18 months, at least about 2 years, at least about 3 years, at least 31. The method of any of embodiments 1-30, wherein the method is about 4 years or at least about 5 years.
[Embodiment 32] The method according to any one of Embodiments 1 to 31, wherein the administration route of the anti-CD70 antibody is intravenous.
[Embodiment 33] The method according to any one of Embodiments 1 to 32, wherein the subject is a human.
[Embodiment 34] The method according to any of embodiments 1 to 33, wherein the anti-CD70 antibody is administered in combination with azacytidine.
[Embodiment 35] The method of any of embodiments 1 to 33, wherein the anti-CD70 antibody is administered in combination with venetoclax.
[Embodiment 36] The method of any of embodiments 1 to 33, wherein the anti-CD70 antibody is administered in combination with azacytidine and venetoclax.
[Embodiment 37] The method of any of embodiments 1 to 35, wherein the anti-CD70 antibody is administered in combination with a fluoroquinolone.
[Embodiment 38] A pharmaceutical composition for the treatment of CD70-expressing cancer, comprising a non-fucosylated anti-CD70 antibody and at least one pharmaceutically compatible component, wherein the anti-CD70 antibody is an anti-CD70 antibody. The composition comprises a heavy chain variable region comprising the three CDRs of SEQ ID NO: 1, a light chain variable region comprising the three CDRs of SEQ ID NO: 2, and an Fc domain, the CDRs of which are defined by the Kabat numbering scheme, 38. A pharmaceutical composition for use in the method of any of embodiments 1-37.
[Embodiment 39] A kit comprising a non-fucosylated anti-CD70 antibody and instructions for using the anti-CD70 antibody in the method of any of embodiments 1 to 37, wherein the anti-CD70 antibody comprises: A kit comprising a heavy chain variable region comprising three CDRs of SEQ ID NO: 1, a light chain variable region comprising three CDRs of SEQ ID NO: 2, and an Fc domain, wherein the CDRs of an anti-CD70 antibody are defined by the Kabat numbering scheme.

Claims (31)

対象においてCD70発現がんを処置する方法に使用するためのフコシル化抗CD70抗体の治療有効量を含む組成物であって、前記方法は、対象においてがん細胞の枯渇をもたらし、対象においてCD70+制御性T細胞(CD70+Treg)の枯渇をもたらさず、抗CD70抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含み、がんが骨髄異形成症候群(MDS)及び急性骨髄性白血病(AML)からなる群から選択される、組成物 A composition comprising a therapeutically effective amount of a non- fucosylated anti-CD70 antibody for use in a method of treating a CD70-expressing cancer in a subject, the method comprising: A heavy chain variable region that does not lead to depletion of CD70+ regulatory T cells (CD70+Tregs) and that the anti-CD70 antibody comprises the three CDRs of SEQ ID NO: 1, where the CDRs of the anti-CD70 antibody are defined by the Kabat numbering scheme; A composition comprising a light chain variable region and an Fc domain comprising the three CDRs of SEQ ID NO: 2, wherein the cancer is selected from the group consisting of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). 抗CD70抗体が、配列番号1のアミノ酸配列に対して少なくとも85%同一のアミノ酸配列を含む重鎖可変領域及び配列番号2のアミノ酸配列に対して少なくとも85%同一のアミノ酸配列を含む軽鎖可変領域を含む、請求項1に記載の組成物The anti-CD70 antibody has a heavy chain variable region comprising an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 1 and a light chain variable region comprising an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 2. 2. The composition of claim 1, comprising: 抗CD70抗体が、配列番号1のアミノ酸配列を含む重鎖可変領域及び配列番号2のアミノ酸配列を含む軽鎖可変領域を含む、請求項1に記載の組成物 2. The composition of claim 1, wherein the anti-CD70 antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 1 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 2. Fcドメインが、抗体依存性細胞傷害(ADCC)、抗体依存性細胞貪食(ADCP)及び補体依存性細胞傷害(CDC)のうち1つ以上を媒介する抗体エフェクタードメインである、請求項1から3のいずれか一項に記載の組成物Claims 1 to 3, wherein the Fc domain is an antibody effector domain that mediates one or more of antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cellular cytotoxicity (CDC). The composition according to any one of the above. Fcドメインが、ADCCを媒介する抗体エフェクタードメインである、請求項1から3のいずれか一項に記載の組成物 4. The composition of any one of claims 1 to 3, wherein the Fc domain is an antibody effector domain that mediates ADCC. FcドメインがヒトFcドメインである、請求項1から5のいずれか一項に記載の組成物 6. A composition according to any one of claims 1 to 5, wherein the Fc domain is a human Fc domain. 抗CD70抗体がボルセツズマブである、請求項1から6のいずれか一項に記載の組成物 7. The composition according to any one of claims 1 to 6, wherein the anti-CD70 antibody is borcetuzumab. 抗体が治療剤にコンジュゲートされている、請求項1から7のいずれか一項に記載の組成物 8. The composition of any one of claims 1-7, wherein the antibody is conjugated to a therapeutic agent. 治療剤が、化学療法剤又は免疫調節剤である、請求項8に記載の組成物9. The composition according to claim 8, wherein the therapeutic agent is a chemotherapeutic agent or an immunomodulator. 治療剤が化学療法剤である、請求項8に記載の組成物 9. The composition of claim 8, wherein the therapeutic agent is a chemotherapeutic agent. 化学療法剤が、モノメチルアウリスタチンE(MMAE)又はモノメチルアウリスタチンF(MMAF)である、請求項10に記載の組成物11. The composition according to claim 10, wherein the chemotherapeutic agent is monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF). 治療剤が免疫調節剤である、請求項8に記載の組成物 9. The composition of claim 8, wherein the therapeutic agent is an immunomodulator. 前記方法が抗CD70抗体の集団を投与することを含み、抗CD70抗体の集団中の各抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含み、抗CD70抗体の集団中の抗CD70抗体の少なくとも50%がコアフコシル化を欠く、請求項1から12のいずれか一項に記載の組成物The method comprises administering a population of anti-CD70 antibodies, each antibody in the population of anti-CD70 antibodies comprising three CDRs of SEQ ID NO: 1, wherein the CDRs of the anti-CD70 antibody are defined by the Kabat numbering scheme. 13. The antibody of claims 1 to 12, comprising a heavy chain variable region, a light chain variable region comprising the three CDRs of SEQ ID NO: 2, and an Fc domain, wherein at least 50% of the anti-CD70 antibodies in the population of anti-CD70 antibodies lack core fucosylation. Composition according to any one of the above. 抗CD70抗体の集団中の抗CD70抗体の少なくとも70%が、コアフコシル化を欠く、請求項13に記載の組成物14. The composition of claim 13, wherein at least 70% of the anti-CD70 antibodies in the population of anti-CD70 antibodies lack core fucosylation. がんがMDSである、請求項1から14のいずれか一項に記載の組成物 15. The composition according to any one of claims 1 to 14 , wherein the cancer is MDS. MDSが再発性又は難治性MDSである、請求項15に記載の組成物 16. The composition according to claim 15 , wherein the MDS is relapsed or refractory MDS. 対象が、MDSのための以前の低メチル化剤(HMA)療法後に処置失敗を経験した、請求項16に記載の組成物17. The composition of claim 16 , wherein the subject experienced treatment failure after previous hypomethylating agent (HMA) therapy for MDS. がんがAMLである、請求項1から14のいずれか一項に記載の組成物 15. The composition according to any one of claims 1 to 14 , wherein the cancer is AML. AMLが再発性又は難治性AMLである、請求項18に記載の組成物 19. The composition according to claim 18 , wherein the AML is relapsed or refractory AML. 対象が、AMLを処置するために2つの以前の処置レジメンを受けた、請求項19に記載の組成物 20. The composition of claim 19 , wherein the subject has received two previous treatment regimens to treat AML. 対象が、AMLを処置するために3つの以前の処置レジメンを受けた、請求項19に記載の組成物 20. The composition of claim 19 , wherein the subject has received three previous treatment regimens to treat AML. がん細胞の少なくとも約0.1%、少なくとも約1%、少なくとも約2%、少なくとも約3%、少なくとも約4%、少なくとも約5%、少なくとも約6%、少なくとも約7%、少なくとも約8%、少なくとも約9%、少なくとも約10%、少なくとも約15%、少なくとも約20%、少なくとも約25%、少なくとも約30%、少なくとも約35%、少なくとも約40%、少なくとも約45%、少なくとも約50%、少なくとも約60%、少なくとも約70%又は少なくとも約80%が、CD70を発現する、請求項1から21のいずれか一項に記載の組成物at least about 0.1%, at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about about 9%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least 22. The composition of any one of claims 1-21 , wherein about 60%, at least about 70% or at least about 80% express CD70. 対象に前記組成物を投与することが、対象に非フコシル化抗CD70抗体を投与する前のがん細胞の量と比較して、少なくとも約5%、少なくとも約6%、少なくとも約7%、少なくとも約8%、少なくとも約9%、少なくとも約10%、少なくとも約15%、少なくとも約20%、少なくとも約25%、少なくとも約30%、少なくとも約35%、少なくとも約40%、少なくとも約45%、少なくとも約50%、少なくとも約60%、少なくとも約70%、少なくとも約80%、少なくとも約90%、少なくとも約95%又は約100%のがん細胞の枯渇をもたらす、請求項1から22のいずれか一項に記載の組成物Administering the composition to the subject may reduce the amount of cancer cells by at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least 23. Any one of claims 1-22, which results in a depletion of cancer cells of about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95% or about 100%. The composition described in Section. 対象に前記組成物を投与することが、対象に脱フコシル化抗CD70抗体を投与する前のCD70+Tregの量と比較して、約20%、約10%、約9%、約8%、約7%、約6%、約5%、約4%、約3%、約2%、約1%又は約0.1%以下のCD70+Tregの枯渇をもたらす、請求項1から23のいずれか一項に記載の組成物Administering the composition to the subject reduces the amount of CD70+ Tregs by about 20%, about 10%, about 9%, about 8%, compared to the amount of CD70+ Tregs before administering the defucosylated anti-CD70 antibody to the subject. 24. Any one of claims 1-23 , which results in depletion of CD70+ Tregs of less than about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1% or about 0.1%. The composition described in Section. ベースラインに対して、非フコシル化抗CD70抗体の投与後に対象において1つ以上の治療効果が改善される、請求項1から24のいずれか一項に記載の組成物25. The composition of any one of claims 1-24 , wherein one or more therapeutic effects are improved in the subject after administration of the non-fucosylated anti-CD70 antibody relative to baseline. 1つ以上の治療効果が、客観的奏効率、応答の持続期間、応答までの期間、無増悪生存期間及び全生存期間からなる群から選択される、請求項25に記載の組成物26. The composition of claim 25 , wherein the one or more therapeutic effects are selected from the group consisting of objective response rate, duration of response, time to response, progression free survival, and overall survival. 与経路が静脈内である、請求項1から26のいずれか一項に記載の組成物 27. A composition according to any one of claims 1 to 26 , wherein the route of administration is intravenous. 対象がヒトである、請求項1から27のいずれか一項に記載の組成物 28. A composition according to any one of claims 1 to 27 , wherein the subject is a human. (i)抗CD70抗体が、アザシチジンと組み合わせて投与される、
(ii)抗CD70抗体が、ベネトクラクスと組み合わせて投与される、
(iii)抗CD70抗体が、アザシチジン及びベネトクラクスと組み合わせて投与される、
(iv)抗CD70抗体が、フルオロキノロン(fluoroquinalone)と組み合わせて投与される、
(v)抗CD70抗体が、アザシチジン及びフルオロキノロンと組み合わせて投与される、
(vi)抗CD70抗体が、ベネトクラクス及びフルオロキノロンと組み合わせて投与される、あるいは
(vii)抗CD70抗体が、アザシチジン、ベネトクラクス及びフルオロキノロンと組み合わせて投与される、
請求項1から28のいずれか一項に記載の組成物
(i) the anti-CD70 antibody is administered in combination with azacytidine;
(ii) the anti-CD70 antibody is administered in combination with venetoclax;
(iii) the anti-CD70 antibody is administered in combination with azacytidine and venetoclax;
(iv) the anti-CD70 antibody is administered in combination with a fluoroquinolone;
(v) the anti-CD70 antibody is administered in combination with azacytidine and a fluoroquinolone;
(vi) the anti-CD70 antibody is administered in combination with venetoclax and a fluoroquinolone; or
(vii) the anti-CD70 antibody is administered in combination with azacitidine, venetoclax and a fluoroquinolone;
29. A composition according to any one of claims 1 to 28 .
CD70発現がんの処置のための医薬組成物であって、非フコシル化抗CD70抗体と、少なくとも1つの薬学的に適合する成分とを含み、抗CD70抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含み、前記組成物が、請求項1から29のいずれか一項に記載ものである、医薬組成物。 A pharmaceutical composition for the treatment of a CD70-expressing cancer, comprising a non-fucosylated anti-CD70 antibody and at least one pharmaceutically compatible component, wherein the anti-CD70 antibody has a CDR having a Kabat number. 29. The composition comprises a heavy chain variable region comprising the three CDRs of SEQ ID NO: 1, a light chain variable region comprising the three CDRs of SEQ ID NO: 2, and an Fc domain as defined by the naming scheme of claims 1 to 29. A pharmaceutical composition according to any one of the above. 非フコシル化抗CD70抗体と、請求項1から29のいずれか一項に記載の組成物を使用するための使用説明書とを含むキットであって、抗CD70抗体が、抗CD70抗体のCDRがKabat番号付けスキームによって規定される、配列番号1の3つのCDRを含む重鎖可変領域、配列番号2の3つのCDRを含む軽鎖可変領域及びFcドメインを含む、キット。
30. A kit comprising a non-fucosylated anti-CD70 antibody and instructions for using the composition according to any one of claims 1 to 29 , wherein the anti-CD70 antibody has a CDR of the anti-CD70 antibody. A kit comprising a heavy chain variable region comprising the three CDRs of SEQ ID NO: 1, a light chain variable region comprising the three CDRs of SEQ ID NO: 2, and an Fc domain, as defined by the Kabat numbering scheme.
JP2022539657A 2019-12-30 2020-12-28 Methods of treating cancer with non-fucosylated anti-CD70 antibodies Pending JP2023508496A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201962954904P 2019-12-30 2019-12-30
US62/954,904 2019-12-30
US202063011906P 2020-04-17 2020-04-17
US63/011,906 2020-04-17
PCT/US2020/067173 WO2021138264A1 (en) 2019-12-30 2020-12-28 Methods of treating cancer with nonfucosylated anti-cd70 antibodies

Publications (2)

Publication Number Publication Date
JP2023508496A JP2023508496A (en) 2023-03-02
JPWO2021138264A5 true JPWO2021138264A5 (en) 2023-12-27

Family

ID=74236285

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2022539657A Pending JP2023508496A (en) 2019-12-30 2020-12-28 Methods of treating cancer with non-fucosylated anti-CD70 antibodies

Country Status (12)

Country Link
US (1) US11820827B2 (en)
EP (1) EP4085073A1 (en)
JP (1) JP2023508496A (en)
KR (1) KR20220133889A (en)
CN (1) CN115605509A (en)
AU (1) AU2020416193A1 (en)
BR (1) BR112022012885A2 (en)
CA (1) CA3166410A1 (en)
IL (1) IL294262A (en)
MX (1) MX2022007974A (en)
TW (1) TW202138388A (en)
WO (1) WO2021138264A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024040194A1 (en) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditioning for in vivo immune cell engineering

Family Cites Families (125)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2036891B (en) 1978-12-05 1983-05-05 Windsor Smith C Change speed gear
US4714681A (en) 1981-07-01 1987-12-22 The Board Of Reagents, The University Of Texas System Cancer Center Quadroma cells and trioma cells and methods for the production of same
US4474893A (en) 1981-07-01 1984-10-02 The University of Texas System Cancer Center Recombinant monoclonal antibodies
ES8504461A1 (en) 1982-04-12 1985-04-16 Hybritech Inc Antibodies having dual specificities, their preparation and uses therefor.
US4486414A (en) 1983-03-21 1984-12-04 Arizona Board Of Reagents Dolastatins A and B cell growth inhibitory substances
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
JPS61134325A (en) 1984-12-04 1986-06-21 Teijin Ltd Expression of hybrid antibody gene
CA1319120C (en) 1985-04-01 1993-06-15 John Henry Kenten Transformed myeloma cell-line and a process for the expression of a gene coding for a eukaryotic polypeptide employing same
CA1282069C (en) 1985-09-12 1991-03-26 Damon L. Meyer Antibody complexes of hapten-modified diagnostic or therapeutic agents
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
EP0247091B1 (en) 1985-11-01 1993-09-29 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US4816444A (en) 1987-07-10 1989-03-28 Arizona Board Of Regents, Arizona State University Cell growth inhibitory substance
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
US5336603A (en) 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
EP0325224B1 (en) 1988-01-22 1996-07-31 ZymoGenetics, Inc. Methods of producing secreted receptor analogs
US4925648A (en) 1988-07-29 1990-05-15 Immunomedics, Inc. Detection and treatment of infectious and inflammatory lesions
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
JP3771253B2 (en) 1988-09-02 2006-04-26 ダイアックス コープ. Generation and selection of novel binding proteins
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5076973A (en) 1988-10-24 1991-12-31 Arizona Board Of Regents Synthesis of dolastatin 3
KR900005995A (en) 1988-10-31 1990-05-07 우메모또 요시마사 Modified Interleukin-2 and Method of Making the Same
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US4978744A (en) 1989-01-27 1990-12-18 Arizona Board Of Regents Synthesis of dolastatin 10
US4879278A (en) 1989-05-16 1989-11-07 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15
US4986988A (en) 1989-05-18 1991-01-22 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptides dolastatin 13 and dehydrodolastatin 13
DE3920358A1 (en) 1989-06-22 1991-01-17 Behringwerke Ag BISPECIFIC AND OLIGO-SPECIFIC, MONO- AND OLIGOVALENT ANTI-BODY CONSTRUCTS, THEIR PRODUCTION AND USE
WO1991000360A1 (en) 1989-06-29 1991-01-10 Medarex, Inc. Bispecific reagents for aids therapy
US5138036A (en) 1989-11-13 1992-08-11 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation and structural elucidation of the cytostatic cyclodepsipeptide dolastatin 14
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5780225A (en) 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
WO1991010737A1 (en) 1990-01-11 1991-07-25 Molecular Affinities Corporation Production of antibodies using gene libraries
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5349053A (en) 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
WO1992005793A1 (en) 1990-10-05 1992-04-16 Medarex, Inc. Targeted immunostimulation with bispecific reagents
ATE160379T1 (en) 1990-10-29 1997-12-15 Chiron Corp BISPECIFIC ANTIBODIES, METHOD FOR THE PRODUCTION THEREOF AND USES THEREOF
EP0564531B1 (en) 1990-12-03 1998-03-25 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
DE69123241T2 (en) 1990-12-14 1997-04-17 Cell Genesys Inc CHIMERIC CHAINS FOR TRANSDUCTING RECEPTOR-RELATED SIGNAL PATHS
EP0580737B1 (en) 1991-04-10 2004-06-16 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
AP257A (en) 1991-04-26 1993-06-03 Surface Active Ltd A method of releasing an antigen from an antibody and methods for their use in diagnosis and therapy.
DE69233482T2 (en) 1991-05-17 2006-01-12 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
JP4124480B2 (en) 1991-06-14 2008-07-23 ジェネンテック・インコーポレーテッド Immunoglobulin variants
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
ATE297465T1 (en) 1991-11-25 2005-06-15 Enzon Inc METHOD FOR PRODUCING MULTIVALENT ANTIGEN-BINDING PROTEINS
ES2227512T3 (en) 1991-12-02 2005-04-01 Medical Research Council PRODUCTION OF ANTIBODIES AGAINST SELF-ANTIGENS FROM REPERTORIES OF ANTIBODY SEGMENTS FIXED IN A PHOTO.
ATE239506T1 (en) 1992-03-05 2003-05-15 Univ Texas USE OF IMMUNOCONJUGATES FOR THE DIAGNOSIS AND/OR THERAPY OF VASCULARIZED TUMORS
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
EP0656064B1 (en) 1992-08-17 1997-03-05 Genentech, Inc. Bispecific immunoadhesins
US5573924A (en) 1992-09-08 1996-11-12 Immunex Corporation CD27 ligand
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US6034065A (en) 1992-12-03 2000-03-07 Arizona Board Of Regents Elucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10
US5410024A (en) 1993-01-21 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
WO1995015982A2 (en) 1993-12-08 1995-06-15 Genzyme Corporation Process for generating specific antibodies
PT1231268E (en) 1994-01-31 2005-11-30 Univ Boston BANKS OF POLYCLONE ANTIBODIES
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
CA2196200A1 (en) 1994-07-29 1996-02-15 Michael Joseph Browne Novel compounds
US5530097A (en) 1994-08-01 1996-06-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory peptide amides
US5504191A (en) 1994-08-01 1996-04-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide methyl esters
US5521284A (en) 1994-08-01 1996-05-28 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides and esters
US5554725A (en) 1994-09-14 1996-09-10 Arizona Board Of Regents Acting On Behalf Of Arizona State University Synthesis of dolastatin 15
US5599902A (en) 1994-11-10 1997-02-04 Arizona Board Of Regents Acting On Behalf Of Arizona State University Cancer inhibitory peptides
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
JP2978435B2 (en) 1996-01-24 1999-11-15 チッソ株式会社 Method for producing acryloxypropyl silane
US6130237A (en) 1996-09-12 2000-10-10 Cancer Research Campaign Technology Limited Condensed N-aclyindoles as antitumor agents
US6239104B1 (en) 1997-02-25 2001-05-29 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear and cyclo-depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
US6075134A (en) 1997-05-15 2000-06-13 The Regents Of The University Of California Glycoconjugates and methods
PT1071700E (en) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
PT1914244E (en) 1999-04-09 2013-07-26 Kyowa Hakko Kirin Co Ltd Method of modulating the activity of functional immune molecules
CA2372053C (en) 1999-04-28 2008-09-02 Board Of Regents, The University Of Texas System Compositions and methods for cancer treatment by selectively inhibiting vegf
US6323315B1 (en) 1999-09-10 2001-11-27 Basf Aktiengesellschaft Dolastatin peptides
US7442776B2 (en) 1999-10-08 2008-10-28 Young David S F Cancerous disease modifying antibodies
US7092985B2 (en) 2000-03-30 2006-08-15 United Devices, Inc. Method of managing workloads and associated distributed processing system
JP2004509612A (en) 2000-06-05 2004-04-02 アバロン ファーマシューティカルズ Oncogene determination and therapeutic screening using characteristic genes
AU2002220265A1 (en) 2000-11-03 2002-05-15 University Of Vermont And State Agricultural College Compositions for inhibiting grb7
US7256257B2 (en) 2001-04-30 2007-08-14 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US20030083263A1 (en) 2001-04-30 2003-05-01 Svetlana Doronina Pentapeptide compounds and uses related thereto
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
WO2004006955A1 (en) 2001-07-12 2004-01-22 Jefferson Foote Super humanized antibodies
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
WO2003026577A2 (en) 2001-09-24 2003-04-03 Seattle Genetics, Inc. P-amidobenzylethers in drug delivery agents
EP1443961B1 (en) 2001-10-25 2009-05-06 Genentech, Inc. Glycoprotein compositions
US7261892B2 (en) 2001-11-27 2007-08-28 Celltech R&D Limited Methods for diagnosis and treatment of epithelial-derived cancers
ES2394018T3 (en) 2001-11-27 2013-01-15 Ucb Pharma S.A. Procedures for the diagnosis and treatment of epithelial cell derived cancers
JPWO2003085107A1 (en) 2002-04-09 2005-08-11 協和醗酵工業株式会社 Genome-modified cells
EP2357006B1 (en) 2002-07-31 2015-09-16 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US7101978B2 (en) 2003-01-08 2006-09-05 Applied Molecular Evolution TNF-α binding molecules
US7662387B2 (en) 2003-02-20 2010-02-16 Seattle Genetics Anti-cd70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
US20080025989A1 (en) 2003-02-20 2008-01-31 Seattle Genetics, Inc. Anti-cd70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
BR122018071968B8 (en) 2003-11-06 2021-07-27 Seattle Genetics Inc antibody-drug conjugate, pharmaceutical composition, article of manufacture and use of an antibody-drug conjugate
US7615211B2 (en) 2004-02-09 2009-11-10 Cbr Institute For Biomedical Research, Inc. CD70 inhibition for the treatment and prevention of inflammatory bowel disease
US8337838B2 (en) 2004-10-15 2012-12-25 Seattle Genetics, Inc. Anti-CD70 antibody and its use for the treatment and prevention of cancer and immune disorders
US7641903B2 (en) 2004-10-15 2010-01-05 Seattle Genetics, Inc. Anti-CD70 antibody and its use for the treatment and prevention of cancer and immune disorders
US20120294863A1 (en) 2004-10-15 2012-11-22 Seattle Genetics, Inc. Anti-CD70 Antibody and Its Use for the Treatment and Prevention of Cancer and Immune Disorders
EP1799262A4 (en) 2004-10-15 2009-10-21 Seattle Genetics Inc Anti-cd70 antibody and its use for the treatment and prevention of cancer and immune disorders
US7875278B2 (en) 2005-02-18 2011-01-25 Medarex, Inc. Monoclonal antibodies against prostate specific membrane antigen (PSMA) lacking in fucosyl residues
AU2006236225C1 (en) 2005-04-19 2013-05-02 Seagen Inc. Humanized anti-CD70 binding agents and uses thereof
EP4026840A1 (en) 2005-07-18 2022-07-13 Seagen Inc. Beta-glucuronide-linker drug conjugates
ES2527961T3 (en) 2005-09-26 2015-02-02 Medarex, L.L.C. Human monoclonal antibodies to CD70
ES2396569T3 (en) 2006-01-17 2013-02-22 Medarex, Inc. Monoclonal antibodies against CD30 that lack fucosyl and xylosyl moieties
MX2009006277A (en) 2006-12-14 2009-07-24 Medarex Inc Human antibodies that bind cd70 and uses thereof.
EP2282773B1 (en) 2008-05-02 2014-01-15 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
WO2012078688A2 (en) 2010-12-06 2012-06-14 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
GB2487551A (en) 2011-01-26 2012-08-01 Rolls Royce Plc Coupling having a threaded interconnector to limit torque
US10196445B1 (en) 2015-03-17 2019-02-05 Bristol-Myers Squibb Company Ipilimumab variant with enhanced ADCC
GB2567613A (en) * 2017-06-16 2019-04-24 Argenx Bvba Treatment for acute myeloid leukaemia
WO2019173692A2 (en) * 2018-03-09 2019-09-12 Agenus Inc. Anti-cd73 antibodies and methods of use thereof
US11873341B2 (en) 2018-03-13 2024-01-16 Tusk Therapeutics Ltd. Anti-CD25 for tumour specific cell depletion
TW202038958A (en) * 2018-12-18 2020-11-01 比利時商阿根思公司 Cd70 combination therapy

Similar Documents

Publication Publication Date Title
KR101092132B1 (en) Treatment of hematologic malignancies associated with circulating tumor cells using chimeric anti-cd20 antibody
US20200206228A1 (en) Combination of an anti-cd19 antibody and a bruton's tyrosine kinase inhibitor and uses thereof
JP2008540447A5 (en)
US20170056496A1 (en) Antitumor combinations containing antibodies recognizing specifically cd38 and cyclophosphamide
US20190076549A1 (en) Combinations of pbd-based antibody drug conjugates with bcl-2 inhibitors
JP2011507932A5 (en)
US20200353077A1 (en) Combinations and uses thereof
JP2020528880A5 (en)
US20220168417A1 (en) Combination therapy with an anti bcma antibody and a gamma secretase inhibitor
RU2010118448A (en) COMBINED THERAPY BY ANTIBODIES ANTI-CD20 TYPE II IN COMBINATION WITH ACTIVE AGENT ANTI-BCL-2
JP2012522513A5 (en)
JP2013539352A5 (en)
CA3101790A1 (en) Anti-cd37 immunoconjugate dosing regimens
JP2015500822A5 (en)
JP2014525926A5 (en)
US11224654B2 (en) Combinations and uses thereof
JP2021501776A5 (en)
JPWO2021138264A5 (en)
JP2021501800A5 (en)
US20210130460A1 (en) Anti-cd19 therapy in combination with lenalidomide for the treatment of leukemia or lymphoma
JPWO2020127503A5 (en)
JPWO2019229677A5 (en)
Routledge Belantamab mafodotin. Antibody-drug conjugate targeting B-cell maturation antigen (BCMA), Treatment of multiple myeloma
JPWO2019183253A5 (en)
BR102022017167A2 (en) ANTI-CD19 THERAPY IN COMBINATION WITH LENALIDOMIDE FOR THE TREATMENT OF LEUKEMIA OR LYMPHOMA