throbber
cancers
`
`Review
`
`FDA-Approved Drugs for Hematological Malignancies—The
`Last Decade Review
`
`Aleksandra Sochacka-Cwikta *, Marcin Maczyriski © and Andrzej Regiec ®
`
`Department of Organic Chemistry and Drug Technology, Faculty of Pharmacy, Wroclaw Medical University,
`211A BorowskaStreet, 50-556 Wroclaw, Poland; marcin.maczynski@umw.edu.pl (M.M.);
`andrzej.regiec@umw.edu.pl (A.R.)
`* Correspondence: aleksandra.sochacka-cwikla@umw.edu.pl; Tel.: +48-7178-406-34
`
`Simple Summary: Hematological malignancies are diseases involving the abnormal production of
`bloodcells. The aim of the study is to collect comprehensive information on new drugs usedin the
`treatment of blood cancers which haveintroducedinto therapy in the last decade. The approved
`drugs were analyzed for their structures andtheir biological activity mechanisms.
`
`Abstract: Hematological malignancies, also referred to as blood cancers, are a group of diseases
`involving abnormalcell growth andpersisting in the blood, lymph nodes, or bone marrow. The
`developmentof new targeted therapies including small molecule inhibitors, monoclonal antibod-
`ies, bispecific T cell engagers, antibody-drug conjugates, recombinant immunotoxins, and, finally,
`Chimeric Antigen Receptor T (CAR-T)cells has improvedtheclinical outcomes for blood cancers.
`In this review, we summarized 52 drugs that were divided into small molecule and macromolecule
`agents, approved by the Food and Drug Administration (FDA)in the period between 2011 and 2021
`for the treatment of hematological malignancies. Forty of them have also been approved by the
`European Medicines Agency (EMA). We analyzed the FDA-approveddrugsby investigating both
`their structures and mechanismsof action. It should be emphasized that the numberof targeted
`drugs wassignificantly higher (46 drugs) than chemotherapy agents (6 drugs). We highlight recent
`advancesin the design of drugs that are used to treat hematological malignancies, which make them
`moreeffective andless toxic.
`
`Keywords: small molecule agents; macromolecule agents; hematological malignancies; FDA; EMA
`
`
`1. Introduction
`
`G checkfor
`updates
`Citation: Sochacka-Cwikia, A.;
`Maczynski, M.; Regiec, A.
`FDA-Approved Drugsfor
`Hematological Malignancies—The
`Last Decade Review. Cancers 2022, 14,
`87. https://doi.org/10.3390/
`cancers14010087
`
`Academic Editor: Marco Picardi
`
`Received: 1 November2021
`
`Accepted: 20 December 2021
`Published: 24 December2021
`
`Publisher’s Note: MDPIstays neutral
`with regard to jurisdictional claimsin
`published mapsandinstitutionalaffil-
`iations.
`
`Hematological malignancies, also known as blood cancers, are diseases characterized
`by the clonal proliferation of blood-forming cells, which occur in blood, bone marrow, or
`lymph nodes. Hematological malignancies include wild range types of leukemia, lym-
`phoma, and myeloma,classified into two types: lymphoid and myeloid [1]. According to
`its mechanism of action, the drugs used for the treatment of hematological malignancies can
`historically be divided into the following groups: deoxyribonucleic acid (DNA)-interactive
`agents, antimetabolites, anti-tubulin agents, and molecular targeting agents such as highly
`specific small molecules and monoclonal antibodies. DNA interactive agents, the old-
`est group of anticancer medications, can be primarily categorized into alkylating agents,
`cross-linking agents, intercalating agents, topoisomerase inhibitors, and DNA-cleaving
`agents [2]. The first alkylating agent approved by the Food and Drug Administration
`(FDA) was chlormethine (mechlorethamine), also called nitrogen mustard. Goodman
`and coworkers described, in 1946, the pharmacological effect of mechlorethamine on
`Hodgkin’s lymphoma, lymphosarcoma, and leukemia [3], which led to this drug being
`registered in 1949 [4]. As a result of work on folic acid antagonists carried out by Farber,
`the next class of drug was developed, i.e., antifolate. In 1948, Farber reported the use
`of aminopterin, which was the 4-amino derivative of folic acid, to treat children with
`CELGENE 2115
`APOTEX v. CELGENE
`IPR2023-00512
`
`Copyright: © 2021 by the authors.
`Licensee MDPI, Basel, Switzerland.
`This article is an open accessarticle
`distributed under
`the terms and
`conditions of the Creative Commons
`
`Attribution (CC BY) license (https://
`creativecommons.org/licenses/by/
`4.0/).
`
`Cancers 2022, 14, 87. https: / /doi.org/10.3390/cancers14010087
`
`https:/ /www.mdpi.com/journal/cancers
`
`

`

`2 of 64
`Cancers 2022, 14, 87
`
`acute leukemia [5]. Methotrexate (amethopterin) replaced aminopterin in the treatment
`of patients in 1953 becauseit has a better therapy-versus-toxicity ratio [6,7]. Then, mer-
`captopurine and fluorouracil were discoveredasthe first structural analogs of purine and
`pyrimidine, respectively. Mercaptopurine was synthesized by Elionetal. in 1952 [8] and
`wasfirst FDA-approved in 1953 [9], while fluorouracil was developed by Dushinskyetal.
`in 1957 [10] and received first approval in 1962 [11]. These drugs were widely used for
`the treatment of both solid and hematological malignancies [12]. Generally, folate, purine,
`and pyrimidine antagonists form oneof the oldest classes of anticancer drugs, i.e., an-
`timetabolites. The next discovered agents for the treatment of hematological malignancies
`were natural plant alkaloids with anti-tubulin activity. Noble and Beerisolated twofirst
`vincaalkaloids,i.e., vinblastine and vincristine, from Catharanthus roseus (L.) G. Don [13].
`Both compoundsreceived extensive clinical evaluation leading to the FDA approvalof
`vincristine in 1963 as therapies for a variety of cancers [14]. Other natural products were
`cytotoxic antibiotics such as bleomycin and doxorubicin. Bleomycin was found in Strep-
`tomycesverticillus by Umezawaetal. in 1962. This antibiotic wasthe first DNA-cleaving
`agentto be registered, in 1973 [15], and can be used to treat malignant lymphomaas well
`as squamouscell carcinomaof the skin, head, and neck [16]. Doxorubicin wasisolated
`from Streptomyces peucetius var. caesius in 1967in Italy [17], and wasfirst FDA-approved in
`1974 [18]. The drug showed anticanceractivity via multiple mechanismsincluding interca-
`lation into DNAandinhibition of topoisomeraseII activity. Doxorubicin was commonly
`used for the treatmentof various hematological malignancies [19]. In 1965, Rosenberg and
`co-workersdiscoveredthat cisplatin, the platinum coordination complex synthesized by
`Peyronefor the first time in 1845 [20], caused inhibition of cellular division [21]. Then,
`cisplatin was entered in trials against a wide range of cancers where it showed potent
`anticancer activity through the cross-linking of DNA. The drug was approved by the
`FDAin 1978 and, since that time, has been usedasa first-line treatmentfor patients with
`leukemia or lymphomas. Currently, it is still one of the most successful anticancer agents
`usedin clinical practice [22]. A milestone for blood cancer treatment wasthe discovery of
`targeted therapy, consisting of the inhibition of molecular targets that are specific molecules
`involved in the growth, progression, and spread of cancer by monoclonal antibodies or
`small selective molecules. The first FDA-approved monoclonal antibody for the treatment
`of hematological malignancies, a genetically engineered chimeric anti-cluster of differentia-
`tion 20 (CD20) antibody, was rituximab. The drug wasregistered in 1997 for the treatment
`of relapsedorrefractory, B-cell, low-grade,or follicular non-Hodgkin’s lymphoma (LG/F
`NHL)[23]. Imatinib wasthefirst small molecule inhibitor (SMI) to be foundto be selective
`against variousprotein tyrosine kinases. It was synthesized by Buchdungerin 1996 and ap-
`proved by the FDA in 2001. The drug wasindicated for patients with chronic myelogenous
`leukemia (CML) [24].
`Thisarticle is an overview of drugs usedin the treatment of hematological malignan-
`cies, which was approved by the FDA from 2011 until 2021. The most recent examples of
`small molecule and macromolecule drugs are detailed, focusing on the initial approval
`date, chemical structure, molecular target, route of administration, indication, and the
`most commonadverseeffects for each agent. Depending on the mechanism ofaction, the
`approved drugsare assigned to two categories: chemotherapy andtargeted agents. In the
`presentreview, the medications containing a new molecular entity, or old active ingredient
`but in a new formulation, are summarized. The drugsreferred to as biosimilars are also
`included. The biosimilars are an important group of biologic medicines which, although
`similar in structure, purity, and function to their reference products, with no meaningful
`differencesin clinical efficacy and safety, increase access to hematologic malignancyther-
`apies by mitigating the treatment costs [25]. Notably, the drugs received supplemental
`indications in the period from 2011 to 2021 but were originally approved before 2011, and
`drugsusedto treat the side effects of cancer treatmentare not includedin this work.
`
`

`

`3 of 64
`Cancers 2022, 14, 87
`
`2. Small Molecule Anticancer Drugs
`2.1. Various Protein Kinase Inhibitors as Anticancer Agents
`Protein kinases are enzymes, whichcatalyse the reversible phosphorylation of proteins.
`This reaction is one of the most important regulatory mechanismsandplaysa crucial role
`in processes suchasthe transduction of external signals andthecell cycle regulation. There-
`fore, protein kinases inhibitors are an important group in need of new drugs, especially
`anticancer drugs. Protein kinase inhibitors are divided into three types. Type I inhibitors
`bind within and aroundthe adenosinetriphosphate (ATP) bindingsite of a catalytically
`active protein kinase, causing inhibition of its phosphorylation. TypeII inhibitors bind to
`a hydrophobic pocket adjacent to the ATP binding site and are usually nonselective. In
`contrast, type III inhibitors bindto allosteric sites, remote from the ATP site, and are highly
`selective [26].
`
`2.1.1. Tyrosine Kinase (TK) Inhibitors
`Tyrosine kinases (TKs) are enzymesthat selectively phosphorylate the hydroxyl groups
`of a tyrosine residue in different proteins using ATP. They havea sharein the regulation of
`most fundamental cellular processes such as growth,differentiation, proliferation, survival,
`migration, and the metabolism ofcells, as well as programmedcell death in response to
`extracellular and intracellular stimuli [27]. The human genomecontainsat least 90 tyrosine
`kinase genes, which codify 58 receptor tyrosine kinases (RTKs) and 32 nonreceptor tyrosine
`kinases (NRTKs) [28]. RTKs are surface transmembrane receptors with kinase activity.
`In the structure of the receptor tyrosine kinases, an extracellular ligand-binding domain
`occurs which is connected to an intracellular catalytic kinase domain byasingle pass
`transmembrane hydrophobic helix [27]. RTKs are not phosphorylated and monomeric in
`an inactive state [29]. Activation by ligand bindingto their extracellular domain results in
`receptors’ oligomerization and autophosphorylation of a tyrosine residue within the kinase
`domain. NRTKsare cytoplasmic proteins that have a kinase domain andvarious additional
`signaling or protein-protein interacting domains[27]. They are activated by intracellular
`signals throughthe dissociation of inhibitors, by recruitment to transmembranereceptors,
`and throughtrans-phosphorylation by other kinases [29]. A large number of RTKs and
`NRIKsare associated with cancers; thus, a significant numberof tyrosine kinase inhibitors
`(TKIs) are currently in clinical development. In the last 10 years, the FDA has approved
`four new drugsfor the treatment of hematological malignancies, which are tyrosine kinase
`inhibitors (Table 1). Among them,there are the agents that target non-receptor Bruton’s
`tyrosine kinase (BTK) or non-receptor Sarcoma(Src) and Abelson (Abl) kinases.
`Ibrutinib, acalabrutinib, and zanubrutinib were originally developed as second-line
`therapy for the treatment of mantle cell lymphoma (MCL), a rare and aggressive type of
`blood cancer. To date, ibrutinib has received 11 FDA approvalssinceit wasfirst regis-
`tered in 2013, amongothers, as a breakthrough therapy for patients with Waldenstrém’s
`macroglobulinemia (WM)and chronic lymphocytic leukemia (CLL), who carry a deletion
`in chromosome17 (17p deletion). In 2019, the FDA approved ibrutinib in combination
`with obinutuzumab, an anti-CD20 monoclonal antibody, as the first non-chemotherapy
`regimen for patients with previously untreated CLL [30]. In the same year, acalabruti-
`nib received approval as the second BTK inhibitor to treat patients with CLL or small
`lymphocytic lymphoma (SLL). This drug can be used as monotherapy or in combina-
`tion with obinutuzumab [31]. The mechanism ofaction of ibrutinib, acalabrutinib, and
`zanubrutinibis the irreversible inhibition of BTK activity by forming a covalent bond with
`a cysteine residue in the BTK active site. This results in blocking B cell antigen receptor
`signaling(i.e., nuclear factor of activated T-cells (NFAT) pathway, nuclear factor kappa-
`light-chain-enhancerof activated B cells (NF-KB) pathway, and mitogen-activated protein
`kinase (ERK) pathway), thus inhibiting the malignantB cells’ proliferation and survival
`(Figure 1) [32-34].
`Bosutinib is a dual inhibitor of Src and Abl kinases that is used as a treatment for
`patients with Philadelphia chromosomepositive (Ph+) chronic myeloid leukemia (CML),
`
`

`

`4 of 64
`Cancers 2022, 14, 87
`
`whoshowresistance or intolerance to previous therapy, including imatinib. The indication
`was extendedin 2017 to include patients with newly diagnosed chronic phase Ph+ CML [35].
`The drug showsactivity against most imatinib-resistant mutants of BCR-ABL, whichis
`a hybrid of a breakpoint cluster region protein (BCR) and Abelson tyrosine kinase (Abl),
`except the mutations T315] and V299. Bosutinib does not inhibit either the receptor tyrosine
`kinase c-Kit (known as mast/stem cell growth factor receptor or CD117) orplatelet-derived
`growth factor receptor (PDGFR) [36]. The drug acts by bindingto the active conformation
`of the kinase domain andinhibiting its autophosphorylation, resulting in a blockade of
`cancercell growth (Figure 1) [37].
`
`RTK
`
`Si
`
`Abl
`
`eyKLE
`[alwal
`PCPNel Mlalilieyicels
`x :
`_—<«$_— such as bosutinib
`suchasibrutinib
`
`
`BTKinhibitor
`
`Nucleus
`
`IP3 >> Coe) > NFAT
`
`SIBIDH
`» Cell growth andproliferation ‘
`
`Cytoplasm
`
`Figure 1. Modeofaction of tyrosine kinase (TK) inhibitors such as non-receptor BTK and Src/AbIin-
`hibitors. BCR: B-cell receptor. RTK:tyrosine kinase receptor. RAF: proto-oncogeneserine /threonine-
`protein kinase. MEK: mitogen-activated protein kinase kinase. ERK: mitogen-activated protein
`kinase. Sre: non-receptor Sarcoma kinase. Abl: Abelson kinase. Rac: Ras-related C3 botulinum
`toxin substrate. JNK: c-Jun N-terminal kinase. SYK: spleen tyrosine kinase. BCAP: B cell adapter
`for PI3K. DAG:diacylglycerol. PKC: protein kinase C. IKK: IkB kinase. NF-«B: nuclear factor
`kappa-light-chain-enhancerof activated B cells. Lyn: tyrosine-protein kinase Lyn. BTK: Bruton’s
`tyrosine kinase. PLC: phospholipase C. IP3: inositol trisphosphate. NFAT: nuclear factor of activated
`T-cells. Created with BioRender.com based on informationin [37,38].
`
`

`

`
`
`
`
`
`
`
`
`
`
`UIpaye[Nqe}stS8nIpJoJapIOAY],“LZOZOFLLOZWoy(Vy)uoHeNstuTUpy3niqpuepoojayyAqpaaorddesroyzrqryut(yf)eseuTySUTSOIA}ayyJOsaINyeay"TLaqeI,
`
`
`
`
`
`
`
`
`
`
`
`ysnoo
`
`
`
`Ttydoynaupaseaiseq
`
`
`
`
`
`
`
`3ddAued3aouatajayspajjqasIaapywoldIpUyeoonyrenoDWamypniysvwalvaassinSUIUN)Peigm4‘ON
`
`
`
`
`
`
`[or’6¢]Jeddn‘yunoojayayeyd12DonuyTeIO1LdYHSw610ZroqaonoNFL“py‘oueyiegqrurynaqnuezLpaseaidap
`‘eruoumnaud°NOWasVSNDINUE
`‘UOTsUTapenK1ovendsouewrouduk=m.610Ae67euryp‘Sutfiag
`
`
`
`‘eoysrerp‘Suismniqo4“ysed
`“etuadoynouI“eruraue‘yuNOD“Ho
`
`
`
`
`JOOWIRN]OLIaUas)
`
`
`
`‘aJePUOTLISIZAIJSAP[OO}}UBAISOUJOJapIO
`
`
`
`
`
`
`
`
`
`
`
`yLterp‘oysepeoH]12Dapuew
`
`‘eaylerp‘ayoepee
`
`
`
`
`
`[zrTr’Te]rANOISHYUOD4oyAdoydusTTeIO1MLd—_CN,Se£102goOle“enauaZeSYqruyniqepeoyZz
`
`
`
`ropucueoraetonnauHels‘enwna]0CMoy(CORAMPAONSysa8prquies
`
`,aongstuo1yDHNN7NEN:AONINOTVO
`
`
`vasneu‘ansryey‘ewoyduréy\if‘Vdd
`eruadoyAd0quiomyyonAooydursqZan
`
`
`
`
`‘an3yey‘eaysreiqT8Dapueyy
`
`ewoydws]
`
`
`
`
`
`‘eaudsAp‘easneu‘euroydurAh]
`
`
`
`
`[FEV0E]—ygnooysex‘uoMDojutestlomecopie[10uaCrN;erozananonerHPL"ulaiaaayqaaynaqy
`
`
`
`ealon!at]-dUT[NGO[SOIDeY|NAO“VINie
`
`
`
`Buon‘erspenprees\4PLOZ199010TZvsn“Tl‘yng
`
`
`
`yeyAroyertdsarsaddn“erutayna’]‘wad
`
`“eruadoyAd0quiomy}onAroydursqYt\_-Ne4
`
`
`‘ewape[eraydiiadoyAd0ydus'Tzi“o
`‘uotyedysuosoroHO
`
`
`
`‘eruraue‘etuadoryneu‘euoydu4]
`
`
`
`
`
`
`‘eruournaudauoz[euIsieyy
`
`
`
`uopyeyerpAyapeuroydurs]
`
`
`eruadoyynou‘eruraue
`“etuadoyAd0quiomy}
`
`
`
`
`
`
`
`
`
`
`
`[Zt-S¥]|‘ZUT}TUIOAsnouasopadjyee)£24goOzzrequadagFMaN“ulJ8ZzUqqruynsogyp
`
`
`‘uredTeurmopqeotuo1yDz19HN:dTINSOd
`‘easneu‘eayzIeiq\5cus‘vas
`
`
` aeruraynoyayHONEALNE102NINZzVSI‘ANHOR
`
`
`
`
`
`
`
`‘BSCULYUOS|AGY“[QV¢‘BSLUD]PUIODIeS10}dad—aI-UOU‘1g,‘BseUL]SUISOIAY$,UOINAGLLG|
`
`
`
`
`
`
`
`
`
`$9JOG
`
`
`
`28‘PL‘@ZOTS4aUDD
`
`
`€
`

`

`6 of 64
`Cancers 2022, 14, 87
`
`
`2.1.2. Multi Kinase Inhibitors
`
`Multi kinase inhibitors are a group of ATP-competitive drugs that target a set of
`structurally related kinases. A single multi-inhibitor is preferred to two single inhibitors
`since drug-drug interactions might occur, changing the metabolism andactivities against
`particular kinases. Multi kinase drugs becomethe second choice when their pharmacoki-
`netic properties are worse. Besides, multi kinase inhibitors are less specific and might
`consequently lead to more side effects. A frequently observed disadvantage duringtreat-
`ment with multi kinase inhibitors is acquired resistance [48]. However, the inhibition
`of several kinases by one drugis useful in anticancer therapy, because oncogenesis and
`cancer growthhaveto be considered as multistep processes that are dependenton various
`signaling pathways(Figure 2) [49]. An overview of FDA-approved multi kinase inhibitors
`is presented in Table 2.
`
`PKC
`
` J
`
`/
`{
`fe @eI-
`
`
`bl
`A
`— =
`Ge Nucleus
`Rac=
`|
`|
`Coo))
`NANANONININIS>
`PS) —> Coa) > (rar
`Cell growth andproliferation
`Protein synthesis
`
`IKK
`
`se
`
`Figure 2. Schematic representation of the signaling pathwaysthat can potentially be inhibited by
`multi kinase inhibitors. BCR: B-cell receptor. PDGFR:platelet-derived growth factor receptor. FLT3:
`FMS-like tyrosine kinase-3. AXL: AXL receptor tyrosine kinase. ALK: anaplastic lymphoma ki-
`nase. VEGFR:vascular endothelial growth factor receptor. FGFR:fibroblast growth factor receptor.
`RET:receptor tyrosine kinase rearranged during transfection. c-Kit: mast/stem cell growth fac-
`tor receptor. TIE2: tunica interna endothelial cell kinase 2. PI3K: phosphatidylinositol 3-kinase.
`PIP2: phosphatidylinositol 4,5-bisphosphate. PIP3: phosphatidylinositol-3,4,5-trisphosphate. PTEN:
`phosphatase and tensin homolog deleted on chromosome ten. PDK: 3-phosphoinositide-dependent
`protein kinase. AKT: protein kinase B. mTORC1: mammalian target of rapamycin complex 1. 4E-
`BP1: 4E-binding protein 1. eIF4E: eukaryotic translation initiation factor 4E. S6K: p70S6 kinase.
`S6: S6 protein. RAF: proto-oncogeneserine/threonine-protein kinase. MEK: mitogen-activated
`protein kinase kinase. ERK: mitogen-activated protein kinase. Sre: non-receptor Sarcoma kinase.
`Abl: Abelson kinase. Rac: Ras-related C3 botulinum toxin substrate. JNK: c-Jun N-terminal kinase.
`CDK:cyclin-dependent kinase. SYK: spleen tyrosine kinase. BCAP:Bcell adapter for PI3K. DAG:
`diacylglycerol. PKC: protein kinase C. IKK: IkB kinase. NF-«B: nuclear factor kappa-light-chain-
`enhancerof activated B cells. Lyn: tyrosine-protein kinase Lyn. BTK:Bruton’s tyrosine kinase. PLC:
`phospholipase C. IP3: inositol trisphosphate. NFAT:nuclear factorof activated T-cells. Created with
`BioRender.com based on information in [37,38,50,51].
`
`

`

`7 of 64
`Cancers 2022, 14, 87
`
`The kinase domain mutationsare the reason for developing drugresistance during the
`treatmentof various types of leukemia. One of the most commongenetic alterations is the
`gatekeeper T315] substitution observed in chronic myeloid leukemia (CML) or Philadelphia
`chromosomepositive (Ph+) acute lymphoblastic leukemia (ALL) and FMS-like tyrosine
`kinase-3 (FLT3)-activating mutations in acute myeloid leukemia (AML). Resistance to tyro-
`sine kinase inhibitors has necessitated the designing of new mutation-resistant inhibitors,
`such as ponatinib, midostaurin, and gilteritinib. Ponatinib is a multitarget inhibitor char-
`acterized by high-affinity and optimized bindingto the active site of the BCR-ABL kinase
`domain, in which the T315 can occur. This mutation is the major reason for inhibition
`access of the drug to the enzyme’s ATP-bindingsite, leading to resistance to first- and
`second-generation tyrosine kinase inhibitors [52]. Ponatinib is effective in the inhibition
`of native and mutant BCR-ABL,receptor tyrosine kinase rearranged during transfection
`(RET), FLT3, tunica interna endothelial cell kinase 2 (TIE2), mast/stem cell growth factor
`receptor (c-Kit), vascular endothelial growth factor receptors (VEGFRs), fibroblast growth
`factor receptors (FGFRs), and platelet-derived growth factor receptors (PDGFRs). Treat-
`ment with ponatinib showssubstantial and durable clinical activity in patients with Ph+
`leukemia with resistance or intoleranceto all other approvedtyrosine kinaseinhibitors [53].
`Adverse events of this therapy are defined as follows: nonhematologic toxic effects such
`as skin disorders(e.g., rash, acneiform dermatitis, and dry skin), constitutional symptoms
`(e.g., arthralgia, fatigue, and nausea), or hematologic—suchas vascular occlusive—events,
`venous thromboembolic events, thrombocytopenia, and neutropenia [52]. The occurrence
`of vascular events during therapy was dependenton the dose of ponatinib, wherein lower
`doses have affected the improvementof the vascular safety profile [54]. Midostaurin
`and gilteritinib are approved drugs for patients with newly diagnosed FLT3-mutated
`AML.Theclinical activity of midostaurin in combination with cytarabine and daunoru-
`bicin-based chemotherapy waspositive for FLT3-activating mutations, such as, primarily,
`in-frame internal tandem duplications (ITD) and missense point mutations in the tyro-
`sine kinase domain (TKD). Moreover, midostaurin inhibits c-Kit (wild type and D816V
`mutant) found in advanced systemic mastocytosis (SM), which includes aggressive sys-
`temic mastocytosis (ASM), systemic mastocytosis with associated hematological neoplasm
`(SM-AHN), and mastcell leukemia [55]. It was found to also be an inhibitor of protein
`kinase C (PKC), platelet-derived growth factor receptors (PDGFRs) alpha andbeta, cyclin-
`dependentkinase 1 (CDK1), spleen tyrosine kinase (SYK), and vascular endothelial growth
`factor receptor-2 (VEGFR-2) [56]. Although midostaurin showsa broad spectrum of an-
`tikinase activity, it is characterized by lacked potency. Gilteritinib, on the other hand,is
`a selective, potent inhibitor of all FLT3-activating mutation types(e.g., ITC, TKD, D835Y,
`double ITD-D835Y) [57]. Furthermore, gilteritinib showsactivity against c-Kit and the
`AXLreceptor tyrosine kinase (AXL, also known as UFO), whichis implicated in FLT3
`inhibitor resistance [58]. The mechanism ofaction of gilteritinib involves bindingto the
`active conformation of FLT3 at the ATP-bindingsite, resulting in reduced proliferation of
`cancercells that overexpress the mutation [59].
`The mutations that confer activation of the intracellular Janus kinase (JAK) signal
`transducer andactivator of transcription (STAT) pathways(e.g., JAK2, V617F, and JAK2
`exon 12) were identified as the most commonin patients with myelofibrosis (MF). Only two
`drugs, namely ruxolitinib and fedratinib, are approved as JAKinhibitors for the treatment
`of MF [60]. Ruxolitinib is a JAK1/2 inhibitor that potently inhibits the proliferation of
`JAK2 V617F-driven Ba/F3cells, resulting in decreased levels of phosphorylated JAK2 and
`signal transducer and activator of transcription 5 (STAT5) [61]. Ruxolitinib provides a
`rapid reduction in splenomegaly, ameliorating debilitating myelofibrosis-related symptoms
`and improving quality of life in patients with MF. The adverse events of ruxolitinib, like
`anemia and thrombocytopenia, were manageable andled to the discontinuation of therapy
`at a low rate [62]. Ruxolitinib is also an effective drug for patients with polycythemia
`vera, which allows for hematocrit control, reducing spleen size, and improving symptoms
`of disease [63]. However, some patients lose response to ruxolitinib and discontinue
`
`

`

`8 of 64
`Cancers 2022, 14, 87
`
`
`treatment over time because developing resistance or intolerance is associated with a
`substantially reduced life expectancy. Fedratinib, an alternative approved JAK inhibitor,
`is potent and selective for JAK2 regardless of its mutational status [64]. Compared with
`ruxolitinib, fedratinib causes a more effective reduction in spleen volume anddisease-
`related symptoms[65].
`
`

`

`
`
`
`
`preyAroyertdsastaddn
`
`
`
`‘Terupajed‘uoysayut
`
`
`
`“eruradApSs1adAy
`
`
`
`‘sisdas‘etuoumnaud
`
`
`
`yuourrredunt[euar
`
`
`
`‘SULMUOA‘easneu
`
`“eruadonnouaaqey
`
`
`
`‘ersperyyie‘er3peAypy
`
`
`
`JOS[@AgT]pasvarout
`
`®HOOfHiv
`
`oO
`
`
`
`‘ayoepeay‘SI}Isoonut“DP
`
`‘eulapa‘eayieipa”
`
`
` S4aUDD 19JO6
`
`
`
`
`
`
`
`
`
`
`
`Ssirarar3s109179asiaAuoneotpuuoTyer}sTUTUIpYyyaSre],aamionxayeqpaaoiddyAueduroDpue3niqjo‘0FouPOTPVHeorpulJOaynoyre[Nda[O/,PMSV/VdaIstaueNpurigaureNdi1aua5N
`
`
`
`
`
`
`
`JOJapsOUTpayeNge}SIs8nIpJoJapsOdU],“LZOZOFLLOTWoy(YA)UoHeNstunUpy3niqpuepoo;ayyAqpoaorddesroyiqryuraseury[NUayyJosanyeay‘7aTqe],
`
`
`
`[oz’69]cAay"asreour140“IXas“out‘SnqqunLAz‘TOAQJ
`
`
`
`
`
`
`[7212]‘erxashd‘anyey‘uredrenutayney[e210“€Ll1dstonTheWy8zseonnaseumreyuLMe\sopry
`
`
`
`
`“VW‘uoneiod10>"[99]‘uonedysuos‘Sunnuoa—-SSOZAOPATNTezervlq6lozeneNYOT3u98]9>)quiperpeqI‘easneu
`
`
`
`[29°29]eruadoyAd0quiomyy‘yumungIzozAreniga,ygvsn‘IN
`vansyey‘SOSCUTLURSURI}eSL1d‘VddVIVdSOX
`
`
`
`“0Nq4eeesisovho0seyyDCD‘,Dd2107equiardasgtNob
`“ured|soqn-“TpHAS‘
`
`
`
`Jeutwopaqe‘uoldeayutLOTSATWan>“gANdOdd“y,ST}IeAON
`
`
`
`‘eaudsAp‘easiereruayna]oNIV:“TIJoorquyJoN
`‘eayelday‘vadDIATANI
`‘ysea"‘ewapa6L0ZF990FZvs
`
`
`
`
`
`
`
`‘aSTeTeUTplopaApyaynoyera8107qWIAAON8Zweyseyeisy
`pajzepas-ao1Aap“,WadoddLdVdaka
`
`“eruraue
`
`‘stxeysida‘uonedysuoozt@UdDAA
`
`PIOpeaAyyeynoVvvd
`
`
`
`
`
`
`
`‘ayepUOTJLIYSISaIJSaplOay}0}JUadaIySOUT
`
`28‘PL‘@ZOT
`
`€
`

`

`
`
`
`
`‘SSO]JUSIOM‘eUapa
`
`eruwosut
`
`
`
`
`
`
`
`
`
`
`aoUatasayspajyqasIaapyworyeorpuyyoanoyreno9;0WamnyonyysVING/VaaistSUNpuergsure]3U3Ua5ON
`
`
`
`
`
`:SYTOAA+‘ujajoiduorlsnyTqV-AO“IAV-UOAT1‘Z-40}daoa110jOeyYAMOIS[eIPayJOpUaIe|MseA:7-WADOAA7,‘BSeUPyautsorA}usa[ds2X,,“|eseuLyyuapuedap-urpA:LHADog,‘Daseuryword
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`‘Md¢“91o}daar10;ejYMOISpaatiap-jo[aqe|d:GYIOdg~10}dadaI10jDejYIMoIBpaatap-jopye[d:VAIOdd,JOydaoa11opeV}YAMOIS[JaoWays/jSeLUDY-9.“aseuPyeWoyduwuA|
`
`
`
`
`
`
`
`
`
`
`
`
`onsedeue:yTV¢‘aseUulyautsor4y10}da0011XV“XV;‘€-OSCUPYUISOIAYAXTY-SW“ELTA¢‘7ASeULYsnuel:ZPWf,‘eoUEIEJaroY}UTUAAISSISnApayJOaINjon}sTestySUOIM,1
`autsoiAy1o}dader:LAY,;‘S10}daoar10j9vyYJMoIspaarrop-jafoye|d‘SYAOAd9‘S10idadar10jDeJYIMOISySeTGOAQY‘SYAOAgy‘S10}daoars0j9v¥yYIMors[eTEyJopuaIe[NsSeA
`
`
`
`
`
`
`
`
`
`
`
`
`[rd'ed]‘AuyedomoujerayduadornOUasOOA,(TO"SOR,TLOZFoquiaseqFTeoeuyredqPUHeUOdv‘:Tg::
`
`
`
`
`
`UTOXNyY[]venaraunyUeruounreogTeIOAVI‘6,LAV\_TX,LL0zOqUIOAONOLSoutoDqrunyyG
`
`9Z'SL“equadoyAs0quioryy“e19ey/:“uones0d.i0
`
`
`
`
`
`
`
`
`yJemard‘ssaurzzrponseygoydursy,2LalHNofHVIN‘adprquie)“Duy
`
`
`
`ayoeproy‘eruournaudoquory4SHASTAHO\vadDISN'TOL
`
`
`Aqp‘ysni‘eaudspeayaeat‘{\
`
`“y8noo‘uoIsnyaenone]eelozArn{1vst
`
`
`euedonnouysou-snsiaA-1Jer5.~Ornztozsendyec“qq‘u0;3unOTIM\,
`
`
`van81yey‘eruadoyneu1.NaTlagey‘sytsoonuterTAVIOE—_
`‘uredN[edo‘eaysrerpa‘uoyedysuoo
`
`
`‘suiseds‘erspehur7“ersyeryyre
`
`‘UpyS‘
`
`
`‘sISOIqUOAWwe\—/vylivxvi
`aseasipmNvsn
`
`
`
`
`
`uonensTunUpyyoSrey,ayeqpeaoiddyAueduroppue3niqjo.
`
`
`
`
`
`
`
`"ToseurysnuelDy6,"7OSeUPy[]29TeTEopudeusdjUTLOT]:ZALLg,WoOHasueyyBurppoSuesseasaseury
`
`derpreo‘uoTsuayIadAPY
`
`
`
`[eurumopae‘amnyrey
`
`
`
`‘ayjeddepaseaisap
`
`v9JOOL
`
`
`
`28‘PL‘@ZOTS4aUDD
`
`
`
`‘quod“TtPTdeL
`
`

`

`11 of 64
`Cancers 2022, 14, 87
`
`2.2. Phosphatidylinositol 3-Kinase (PI3K) Inhibitors as Anticancer Agents
`Phosphatidylinositol 3-kinases (PI3Ks) are a family oflipid kinases that phosphorylate
`phosphoinositides at the 3-hydroxyl group ofthe inositol ring that can be used to generate
`phosphatidylinositol 3,4,5-trisphosphate. Amongthese, several classes have been identified
`and characterized by different primary structures and substrate specificities. Class I PI3Ks
`are heterodimers and divide into two groups, IA andIB. Class IA PI3Ksare activated by
`a wide rangeof receptor tyrosine kinases (RTKs) and are frequently implicated in cancer,
`while class IB PI3Ks are activated by G-protein-coupled receptors [77]. Structurally, class
`IA PI3Ksexists in three isoforms («, 8, and 5) and class IB PI3Ksin one isoform (vy). Class
`II PI3Ks are monomeric proteins that consist of three isoforms («, 8B, and 5), whereas
`class III PI3Ks are only one heterodimer composedof a catalytic (Vps34) and regulatory
`subunit [78]. PI3K-related kinases, which can be includedasclass IV of PI3K, are a group of
`protein kinases with structural similarity to PI3K, but withoutthelipid kinase activity. This
`group includes a mammalian target of rapamycin (mTOR), DNA-dependentprotein kinase
`(DNA-PK), ataxia telangiectasia mutated gene product (ATM), and ataxia telangiectasia
`and Rad3-related gene product[79]. The dysregulation of the phosphatidylinositol-3 kinase
`pathway, especially abnormal activation, is one of the most frequently observed in blood
`cancers and an importanttarget of selective anticancer therapies. The three inhibitors of
`PI3K havereceived market approval since 2011 (Table 3). Idelalisib, a first-in-class inhibitor
`of PI3K-5, and the following inhibitors, i.e., copanlisib and duvelisib, directly reduce the
`proliferation and survival of malignant B-cell leukemia and lymphomacells (Figure 3).
`Hence, they were approved by the FDA for the treatment of different types of leukemia
`and lymphoma[80-82]. Duvelisib is a first-in-class dual inhibitor of PI3Ks dueto the fact
`that it also inhibits PI3K-y activity, which leadsto a reductionin the differentiation and
`migration of various components of the cancer microenvironment, such as T helpercells
`and M2 tumor-associated macrophages[82]. It is worth noting that the route of copanlisib
`administration as intermittent intravenousinfusions lead to weaker gastrointestinal toxicity
`compared with the oral treatmentof idelalisib [83].
`
`RTK
`
`‘oT
`\
`om
`O- EK IS
`a
`[ae] 2+ Ca] e+ fr]mTORC1
`x *
`
`4E-BP1
`S6K
`ky
`eg)
`
`Protein synthesis
`
`PI3Kinhibitor
`
`PTE CMe(irliye)
`
`Nucleus
`
`eau
`
`Cell growth andproliferation
`
`Cytoplasm
`
`Figure 3. Mechanism of action of PI3K inhibitors. RTK: receptor tyrosine kinase. PI3K: phos-
`phatidylinositol 3-kinase. PIP2: phosphatidylinositol 4,5-bisphosphate. PIP3: phosphatidylinositol-
`3,4,5-trisphosphate. PTEN: phosphatase and tensin homolog deleted on chromosometen. PDK:
`3-phosphoinositide-dependentprotein kinase. AKT: protein kinase B. mTORC1: mammaliantarget
`of rapamycin complex 1. 4E-BP1: 4E-binding protein 1. eIF4E: eukaryotic translation initiation factor
`4E. S6K: p70S6 kinase. S6: S6 protein. Created with BioRender.com based on information in [51,79].
`
`

`

`‘eydyeaseury-¢jonsourApyeydsoyd:0-yeId¢
`
`
`
`
`ay}Josamyea,j*¢apqey, 28‘PL‘@ZOT
`
`
`
`
`
`
`
`
`
`
`
`
`Sse1819199779asioAwonestpuuoTyer}sTUTUIpYyyosiey,aamionxayeqpeaoiddyAuedwodpue3niqjo0FOUPOFAPVryeorpuyjoaynoyTe[Ma]/0O/PNASV/VdaIstauIeNpurigWLaUasNauIeN]
`
`
`
`
`
`
`
`snipJOJapIOay,“LZOZOF[LOZWoy(Yq)uoVeNstUUpy3niqpuepooayyAqpaaoiddesioyiqryut(speq)aseury¢-[oysour;Apyeydsoyd
`
`*0Nane/‘}|I:-eslerohtedAlssepnoHTIOsnouaarauy‘,PMEldCy“7™21gwequiaidasgE©sfeonnaveueyg——=ESHUPODcI
`
`
`
`
`
`
`
`
`[68'88]paseadap‘sisdasondvoudunty[tO1SAEId7HN,SyLozAm€z‘SADUBIDSPeTIDESHTPIOPL©
`
`
`
`
`
`S8'T8‘eayslierp‘erurauepuroTeIO“o-SIEId8LOztaquiaydas$7“Duy‘UaySeIIA,arsteandIonAd0yduré’Ife]
`
`
`
`
`
`
`[ow'ee]easneu‘erxarkdSepa]eyzAMeld“VW“VW‘weypaoNIstaan
`
`
`
`
`
`,‘yunod[rydosyneau1._“VINE‘AWDJ94soJ“OUT
`prusdonneu‘suonDosUteuroydurATuorsnjure-MeldSmApesordde
`
`
`
`
`
`‘enupuardisiayieddymounCOPLZtoquiaydaggyiyVS‘vO
`
`
`
`‘erxaidd‘eruadorynau“yorN|‘VddOITYAAZ
`
`
`“eruadoyAo0quionyannon)‘I‘VadVULMIdOD
`
`ewoydus]&___‘SupTUOAremowod1207API61vsn.
`
`
`‘VddarleguyeeayJakeg
`
`
`pueouturyepayeaayaOG
`
`
`“ser‘eaudsAp‘T[ryo:SNOH
`unio.
`
`“erutaoA[81adAyCh
`
`Nl>vs
`
`saseuruesuesy
`ajeyredse
`
`
`
`‘easneu‘eoyselqd
`
`
`
`vansryey‘SuTTUIOA
`
`
`
`“eruounaud‘aysepeoyN=/
`
`‘etuadonan:
`
`VdOOI1TV
`
`‘a}EPUOTWIISIZAISAP[OO}JUIIAISOUJOJapIOUTpayepnqe}st
`
`V9JOCL
`
`S4aUDD
`
`
`
`
`
`
`
`
`
`“eureaseury-¢jousourtApyeydsoyd:A-yeldz“eHepaseuny-¢[oysourjApyeydsoyd:9-yETd1
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`

`13 of 64
`Cancers 2022, 14, 87
`
`2.3. Various Enzymes Inhibitors as Anticancer Agents
`Enzymesare organic high-molecular-weight molecules that catalyze the synthesis or
`degradation reaction of a specific enzyme’s substrate. Enzymeinhibitors bind to the
`enzyme, resulting in disruption of the normal format

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket