EXCLI J EXCLI Journal 1611-2156 Leibniz Research Centre for Working Environment and Human Factors 2022-5653 10.17179/excli2022-5653 Doc146 Review article Bortezomib advanced mechanisms of action in multiple myeloma, solid and liquid tumors along with its novel therapeutic applications Alwahsh Mohammad * 1 2 3 Farhat Joviana 4 Talhouni Shahd 1 Hamadneh Lama 1 Hergenröder Roland 2 Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany Institute of Pathology and Medical Research Center (ZMF), University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, PO Box 127788, United Arab Emirates *To whom correspondence should be addressed: Mohammad Alwahsh, Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan, E-mail: M.alwahsh@zuj.edu.jo 16 01 2023 2023 22 146 168 02 12 2022 12 01 2023 Copyright © 2023 Alwahsh et al. 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (http://creativecommons.org/licenses/by/4.0/) You are free to copy, distribute and transmit the work, provided the original author and source are credited.

This article is available from https://www.excli.de/vol22/excli2022-5653.pdf

Bortezomib (BTZ) is a first-in-class reversible and selective proteasome inhibitor. It inhibits the ubiquitin proteasome pathway that leads to the degradation of many intracellular proteins. Initially, BTZ was FDA approved for the treatment of refractory or relapsed multiple myeloma (MM) in 2003. Later, its usage was approved for patients with previously untreated MM. In 2006, BTZ was approved for the treatment of relapsed or refractory Mantle Cell Lymphoma (MCL) and, in 2014, for previously untreated MCL. BTZ has been extensively studied either alone or in combination with other drugs for the treatment of different liquid tumors especially in MM. However, limited data evaluated the efficacy and safety of using BTZ in patients with solid tumors. In this review, we will discuss the advanced and novel mechanisms of action of BTZ documented in MM, solid tumors and liquid tumors. Moreover, we will shed the light on the newly discovered pharmacological effects of BTZ in other prevalent diseases.

bortezomib proteasome inhibitor multiple myeloma solid tumors liquid tumors
Introduction

Proteasome is a large protein complex responsible for intracellular proteolysis stabilization and regulation of the protein quality control by destroying misfolded or aggregate-prone damaged protein (Collins and Goldberg, 2017[28]; Davis et al., 2021[32]; Majumder and Baumeister, 2019[95]; Mao, 2021[98]). Proteasome is also used by eukaryotic cells to maintain myriad cellular pathways by a gradual key proteins excretion mechanism (Bard et al., 2018[8]; Becker et al., 2019[12]; Dong et al., 2019[36]; Fritze et al., 2020[42]; Majumder et al., 2019[96]). Specifically, 26S proteasome is responsible for the cytoplasmic, nucleic and superficial proteolysis of approximately all organelles (Ding et al., 2017[34], 2019[35]; Worden et al., 2017[160]). Within the 26S proteasome, a 19S regulatory particle (RP) exhibits the degradation signal and unfolding process of the target protein-substrate (Wehmer et al., 2017[158]; Zhu et al., 2018[176]). The 20S core particle (CP) constitutes the catalytic core of the 26S proteasome which dissociates the unfolded polypeptide into shorter peptides or amino acids (de la Peña et al., 2018[33]; Mendes et al., 2020[102]). However, the free 20S proteasome complex is capable of retaining basal proteolytic activity for substrates with an unstructured or unfolded stretch (Boughton et al., 2020[17]; Clague et al., 2015[27]; Longworth and Dittmar, 2019[92]; Oh et al., 2018[119]; Yau et al., 2017[167]).

In the mid-1990s, Adams et al. thought that if the proteasome was responsible for eliminating damaged or harmful proteins in the cell, it might also be removing beneficial ones which originally prevent the acquisition of complex diseases such as cancer (Wang et al., 2017[157]). Consequently, experimental studies have proved the close connection between proteasome dysfunction and cancer disease progression (Schweitzer et al., 2016[133]). This pathophysiological association helped researchers to develop an entirely new class of anticancer drugs called proteasome inhibitors such a bortezomib (BTZ) (Jakubowiak et al., 2011[64]).

BTZ was originally known as PS-341 and developed by ProScript, a biotech company, to treat muscle weakness and muscle loss associated with AIDS and muscular dystrophy (Jakubowiak et al., 2009[65]). The National Cancer Institute (NCI) assessed if PS-341 could be effective in a variety of cancer cell lines (Niu et al., 2021[117]). Researchers found that PS-341 significantly reduced cell growth in many different types of cancer cells by slowing down the action of the proteasome leading to the destruction of key proteins that help cells fight off cancer (Hallberg and Palmer, 2013[52]). BTZ was seen to potentially limit the chymotrypsin-like proteasomal activation by reversibly interacting with the β5 subunit of the 20S proteasome leading to a complete inhibition of proteasomal activity followed by the accumulation of poly-ubiquitinated proteins in cells (Yau et al., 2015[166]). In 2003, US FDA approved BTZ for the treatment of relapsed and refractory Multiple Myeloma (MM) (Adams, 2014[1]). MM is an incurable cancer of the blood that affects approximately 14,000 patients in the US, annually (Adams, 2014[1]). BTZ was also applied in relapsed mantle cell lymphoma and diffuse large B-cell lymphoma (Vesole et al., 2015[154]). In 2005, US FDA fully approved BTZ use as a second-line MM therapy, and as a first-line therapy for patients with newly diagnosed MM after only three years (Korde et al., 2015[77]). Currently, BTZ is being studied for use in a wide variety of blood cancers and solid tumors (Richardson et al., 2014[129]).

In the present review, we will discuss the recently discovered advances of BTZ mechanisms of action in MM, solid and liquid tumors and diseases other than cancer.

Advanced Mechanism of Action of BTZ in MMIntrinsic pathways activation

Calreticulin (CALR) is a chaperone protein found in the endoplasmic reticulum (ER), the cytoplasm, and at the outer surface of the cell. CALR is involved in the proper folding of newly formed proteins and maintenance of calcium ions levels and gene activity control, stabilization of cell growth and division, proliferation, migration and adhesion mechanisms in addition to the regulation of programmed cell death known as apoptosis (Gold et al., 2010[46]; Malcovati et al., 2014[97]; Michalak et al., 2009[106]). CALR pathway activation constitutes the base of a successful BTZ treatment for MM cells. Indeed, CALR processing is initiated through the phosphorylation of eukaryotic initiation factor 2α (eIF2α) by the ER stress kinase eIF2α kinase-3 (EIF2AK3), best known as PRKR-like endoplasmic reticulum kinase (PERK) which induces an integrated response stress (Hopfner and Hornung, 2020[56]; Nangalia et al., 2013[114]). In response to chemotherapy inducers such as BTZ, this phosphorylation event occurs downstream of a general inhibition of DNA-to-RNA transcription. High-dose BTZ is proved to reduce global RNA synthesis in osteosarcoma cells, suggesting that this can also occur in MM cells (Kroemer and Zitvogel, 2021[78]). However, this hypothesis is subject to further investigations.

Moreover, BTZ is characterized by immune-stimulatory effects which express heat shock protein 90 on MM cells, easing their recognition by dendritic cells (DCs) (Spisek et al., 2007[139]). Thus, intracellular proteins will flow toward the cell surface, marking the Immunogenic Cell Death (ICD). Consequently, CALR of the endoplasmic reticulum ER is released from the ER lumen to the plasma membrane surface. There, CALR initiates the phagocytosis of dying cells by DCs through the “eat-me” signal. This CALR-dependent ICD emphasized on BTZ ability to drive CALR toward the surface of human or mouse MM cells leading to the phagocytosis of these cells (Gulla et al., 2021[50]). Hence, achieving an optimal therapeutic efficacy of BTZ against MM cells is suggested to depend on CALR activation and the patient's immune system. In vivo, MM cells were only responsive to BTZ treatment in the immunocompetent setting rather than in immunodeficiency state. But CALR deterioration in MM cells limited the efficacy of BTZ against MM tumors even in immunocompetent mice (Galluzzi et al., 2017[43]). Also, when BTZ-killed murine MM cells were injected into immunocompetent mice, MM cells were unable to grow 1 week post-injection (Galluzzi et al., 2017[43]). This vaccination effect was reduced when the CALR gene was inactivated in MM cells.

In addition to CALR, STING pathway also reflected a major factor behind cancer cells responsiveness toward BTZ. STING is responsible for cell death induction and release of cancer cell antigens. STING activation has been proved to enhance cancer antigen presentation, contribute to the priming and activation of T cells, and facilitate the trafficking and infiltration of T cells into tumors in order to kill cancer cells (Zhu et al., 2019[175]). In fact, STING drop-out prevented the activation of IFNA1, IFNB1, and CXCL9 gene transcription by BTZ and abolished the capacity of BTZ-treated human MM cells to stimulate CD4+ EM and CD8+ EM cells in co-cultured immune cells. When combined with a synthetic STING agonist, ADUS-100, BTZ responses were potentiated in a mouse MM model through a fortified infiltration of the tumors by T lymphocytes and an increased phosphorylation of TBK1 in vitro. These discussed additive effects were minimized when STING was limited in mouse MM cells or in immune-deficient mice (Montes de Oca et al., 2021[110]).

So, CALR and STING pathways together account for a complete BTZ pharmacological activity. STING by translocating to the ER, inter-locates with the CALR pathway. Recent trials showed that STING suppression did not influence BTZ-induced CALR exposure. While in vivo, CALR knockout prevented the BTZ-mediated induction of interferon stimulated genes in MM mouse model (Tallarida, 2011[144]). The need to focus on the importance of CALR in cytosolic DNA-cGAS-STING-TBK1-type-1 interferon pathway generated a new hypothesis for investigation through the in vitro setting. Challenge also apply on specifying the concentration at which CALR deficiency can inhibit this pathway taking into account the potential of cancer cells to escape immune-surveillance by suppressing the CALR exposure pathway at multiple levels (Tallarida, 2011[144]). MM cells can succeed to develop such pathway deficiencies as a result of immune-selection, particularly after recurrence of cancer following BTZ-based therapeutic regimens.

Anti-tumor mediators and intrinsic factors expression

Several anti-tumor mediators have been demonstrated to be involved in the control of growth, progression, and dissemination of MM (Kuku et al., 2005[79]). Co-culturing of BTZ-treated human MM cells with DCs and T lymphocytes induced a high release of CD4+ effector memory (EM) cells, CD8+ EM cells and CD8+ T EM cells re-expressing CD45RA (TEMRA). In fact, the production of these immunity boosters was not seen when BTZ treated MM cells were removed from the co-culture or when BTZ alone was added to the co-culture of DCs and T cells (Zitvogel et al., 2016[178]).

Concomitantly, BTZ efficacy was correlated with the activation of particular expressed genes. In CALR-expressing (but not in CALR-deficient) MM, BTZ stimulated 90 immune-related genes that composed the “ICD signature”. Among them, 57 interferon genes responsible for activating a type-1 interferon response mediated by interferon alpha1 (IFNA1), interferon beta1 (IFNB1) and Chemokine (C-X-C motif) ligand 9 (CXCL9, a ligand acting on the receptor CXCR3 on T cells to attract them into the tumor bed) upregulation in in vitro MM cells, correlating with the clinical outcome of MM patients treated with BTZ-based regimens (Galluzzi et al., 2020[44]). This type-1 interferon response following BTZ administration caused a high release in cytosolic DNA in the form of micronuclei, accumulation of the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS), activation of transmembrane protein 173 (TMEM173, best known as STING) and activation of TANK-binding kinase 1 (TBK1) phosphorylation. Data confirms that BTZ is able to induce a type-1 interferon response usually linked to ICD and has been known as “viral mimicry” (Sistigu et al., 2014[136]). Like other ICD-inducing drugs, BTZ stresses and kills MM cells mimicking the viral infection process to the degree that high levels of type-1 interferons are activated. Recently, BTZ exhibited its potential to upregulate a key cellular miRNA involved in T cell function known as miR-155. This activation resulted in downregulating of BTZ targets which are cytokine signaling 1 (SOCS1) and inositol polyphosphate-5-phosphatase (SHIP1), normally classified as negative endogenous proteins, leading to a suppressed PD-1-mediated T cell exhaustion (Renrick et al., 2021[128]). BTZ confirmed its novel lymphocyte-stimulatory interactions as a crucial factor towards lessening the immunosuppressive actions of tumor on antitumor T cell functions and combining BTZ with other immunotherapies to shrink tumor microenvironment's potential maximally and obtain a full BTZ efficiency (Franchi et al., 2021[41]).

BTZ Anti-Cancer Efficacy in MM

Novel BTZ mechanisms of action led researchers to evaluate its clinical efficacy when used solely or as a part of an anti-tumor regimen.

Recent clinical findings proved that MM patients are living longer than those diagnosed 10 years ago because of the availability of new targeted drugs like BZT (Velcade®). In practice, BTZ single therapy prolonged the median overall survival (OS) and the restricted mean survival time (RMST) to a total of 33.9 and 42.9 months, respectively, compared to a lower finding in the chemotherapy group as 27.9 and 38.4 months, respectively (Tutt et al., 2021[150]). However, Velcade® has shown a greater effectiveness when used in combination with two, three, or four other anticancer drugs in patients newly diagnosed with MM. Nowadays, BTZ containing regimens are the guideline treatment option for MM patients. The combination therapy with BTZ, lenalidomide and dexamethasone (VRd) constitutes the first-line regimen for relapsed/refractory as well as newly diagnosed MM in terms of its high impact on overall survival (Ibata et al., 2016[62]). In a three-drug study, 85 percent of patients responded well to the treatment plan whereas 40 percent of patients experienced remission after one to two years. Also, triple use of BTZ, cyclophosphamide and dexamethasone (VCD) achieved an 88 %, 63 % and 89 % of overall response rate (ORR), progression-free survival (PFS) and overall survival, respectively (Choueiri et al., 2021[25]). BTZ, lenalidomide, and dexamethasone VRd marked a greater overall response rate (ORR), progression-free survival (PFS) and overall survival rates of 98 %, 85 % and 95 %, respectively (Choueiri et al., 2021[25]; Okazuka et al., 2020[120]). In BOSTON and OPTIMISM studies, combining BTZ and dexamethasone with selinexor or pomalidomide provided MM patients with 13.93 months and 11.20 months survival free periods following treatment (Parsons et al., 2020[121]; U.S. FDA, 2022[153]). Similar success occurred with a four-drug combination where 96 percent of patients responded to the treatment and 39 percent went into remission (Wakelee et al., 2021[156]).

In contrast, Cavo et al. proved that hematopoietic stem-cell transplantation (HSCT) in transplant eligible patients can extend patients' PFS to 56.7 months compared to a PFS of 41.9 if those patients will receive BTZ-melphalan-prednisone (VMP) treatment (Cavo et al., 2020[21]). Hence, BTZ used as part of the VRd regimen imposed its greater value as a consolidation therapy, where a 58.9 months' PFS was achieved compared to a 45.5 months' PFS in absence of a consolidated use of BTZ regimen (Cavo et al., 2020[21]). Despite the therapeutic effectiveness of BTZ regimens, patient adherence to a weekly BTZ dose of 1.0 mg/m2 for a 48 weeks' treatment period allowed a maximal successful treatment outcome (Ibarra et al., 2021[61]). Sometimes, severe adverse events may occur without regimen's dose reduction during consolidation/maintenance phase. Recent trials approved that at least six 28-day cycles of subcutaneous BTZ (1.3 mg/m2 on days 1 and 15), lenalidomide (10 mg on days 1-21) and dexamethasone (40 mg on days 1, 8, 15 and 22) regimen will be associated with an overall response rate and complete response (CR) rate equaling 100 and 43.8 %, respectively. Also, PFS and OS rates at 2.5 years were 66.6 % and 77.3 %, respectively where grade 3 or 4 hematologic or non-hematologic adverse events were not documented (Ibata et al., 2016[62]).

In delayed transplantation scenarios, BTZ also proved its efficacy. VRd regimen prolonged PFS and OS for a total duration of 43 months, 75 months respectively compared to 30 months PFS and 64 months OS values when lenalidomide and dexamethasone (Rd) were used in absence of BTZ. Moreover, a larger fraction of patients in VRd group 16 % (34/216) compared to Rd group 8 % (18/214) achieved a better ORR equal to 82% rather than 72 %, respectively (Durie et al., 2017[38]). Yet, toxicological risks were marked by higher proportions of adverse events of grade 3 or higher reported in (82 %) of patients treated with VRd and (75 %) in patients subject to Rd (Durie et al., 2017[38]). Consequently, researchers formulated a more tolerated version of the RVd known as RVd lite mainly indicated to be used for patients ineligible to transplantation. ORR was achieved by 86 % accompanied with peripheral neuropathy and was reported in 31 (62 %) patients with only 1 patient experiencing grade 3 symptoms (O'Donnell et al., 2018[118]).

In patients with relapsed and refractory cases of MM, prolonged or enhanced efficacy of BTZ are needed. Remarkably, in multiple myeloma, patients receiving BTZ were able to achieve complete responses associated with longer life expectancy. At the same time, chromosomal mutations were seen to diminish the efficacy of many anticancer drugs and hence worsen cancer patient prognosis and outcome of multiple myeloma treatment such as del (17q13) while del (13q14), amp (1q21), t (4,14), t (14,16) allowed BTZ to overcome poor prognosis (Grosicki et al., 2020[49]). Accordingly, immunoglobulin subtypes IgA and mainly IgG are considered direct biomarkers for driving resistance toward suboptimal response and VRd treatment failure (Keruakous et al., 2021[73]). Bahlis et al. found that venetoclax plus daratumumab/dexamethasone (VenDd) and VenDd with BTZ (VenDVd) produced high rates of durable responses in those patients (Bahlis et al., 2018[7]). BTZ was also evaluated in combination with mitoxantrone, vincristine, pegaspargase and dexamethasone where of the 10 patients enrolled, eight (80 %) achieved a complete remission (CR) or complete remission with incomplete recovery (CRi). In addition to the occurrence of grade 3 or higher infections in four out of 10 patients, and other toxicities commonly associated with BTZ were not seen (August et al., 2020[6]). As BTZ doses are usually administered in cycles, the risk of cellular resistance acquisition in BTZ treated patients remains a major challenge for its clinical usage in practice. Hence, assessing the resistance mechanism of methylome in neuroblastoma cells following BTZ therapy was performed through genome wide methylation process. This cellular shift bypassed the primary anticancer activity of BTZ, thus expressing a proliferative phenotype proportional to the number of treatments administered. Similar cellular effects were not found with lenalidomide treatment nor with non-treated cells cultured under the same experimental conditions. This phenomenon seems to be directly correlated with BTZ treatment (Łuczkowska et al., 2021[93]). Overall, the implementation of new and adjuvant therapies is boosting the trend towards individualizing treatment as well as ameliorating patients' outcomes in case of a high residual risk of recurrence following primary treatment (U.S. FDA, 2018[151], 2020[152]; Sabbah et al., 2020[132]; Tutt et al., 2021[150]; Wakelee et al., 2021[156]).

BTZ Implementation in Solid Tumors

BTZ was preliminarily approved for the treatment of MM. Nowadays, BTZ constitutes one of the widely used anticancer agents in other solid tumor types (see Table 1(Tab. 1); References in Table 1: Adams, 2001[2]; Alsahafi et al., 2019[3]; Bashraheel et al., 2020[9]; Benvenuto et al., 2021[13]; Bielskienė et al., 2015[14]; Bray et al., 2018[18]; Carbone et al., 2020[20]; Cerruti et al., 2017[22]; Chow, 2020[26]; Dai et al., 2020[30]; Harsha et al., 2020[55]; Hou et al., 2019[57]; Joshi and Broughman, 2021[68]; Kitamura et al., 2020[75]; Laszlo et al., 2019[80]; Lee et al., 2018[83], 2021[82]; Li et al., 2018[84]; Lo Nigro et al., 2017[91]; Montagnani et al., 2017[109]; Muenchow et al., 2020[111]; Nadhan et al., 2020[112]; Pettersson et al., 2013[122]; Raza et al., 2017[127]; Su et al., 2021[140]; Takács et al., 2020[143]; Taniguchi and Karin, 2018[145]; Tsumagari et al., 2018[149]; Wu et al., 2018[161]; Yang et al., 2018[164]; Zhang et al., 2018[171], 2019[170]).

Ovarian cancer

BTZ proved its potential to slow the growth and shrink the size of ovarian tumor when combined with an IKK inhibitor named as Bay 117085. BTZ anticancer property was marked through decreased tumor levels of S536P-p65 NFκB in addition to a minimized recruitment to IL-8 promoter in tumor tissues, alleviating levels of IL-8. Further investigations specified that IKK inhibition limits the IL-8 production and potentiates BZ effectiveness in reducing ovarian tumor growth in vivo and in turn improving patient response to treatment (Huang et al., 2014[58]). Also, BTZ was seen to grant docetaxel additional effects on ovarian cancer cells through the stabilization of apoptotic proteins like Apaf-1, inhibiting the degradation of cytosolic cytochrome c released by docetaxel, stimulating intrinsic apoptosis and initiating cancer cell death. These favorable mechanisms correlate with a better cellular sensitivity toward docetaxel, leading to a proper chemo-resistance control (Weyburne et al., 2017[159]).

Bladder cancer

BTZ has been studied in bladder cancer and failed a Stage II clinical trial. Assessment of its response rate, safety, and toxicity, progression-free and overall survival exhibited its ability to be safe yet not a drug of choice as a second line therapy due to the absence of anti-tumor activity (Mirabella et al., 2011[107]; Radhakrishnan et al., 2010[123]). Currently, 3D drug screening in bladder cancer cell lines highlighted BTZ among the “very active” compounds across the 17 bladder cancer cell lines tested, on the basis of its drug sensitivity score 3 (DSS3) (Rajkumar, 2016[124]). So, futuristic application of experimental technologies is needed in order to rate the importance of BTZ possible contribution as a direct therapy in bladder cancer treatment.

Cervical cancer

Till now, there is no specific study of BTZ on cervical cancer cells but some secondary trials were implemented. In cervical cancer, cells stabilize low levels of intrinsic p53 in function to its rapid proteasomal degradation by E6 and E6-AP proteins (Wustrow et al., 2013[162]). Hence, p53 cellular reactivation mechanism is exhibited either by inhibiting E6 protein at transcriptional and translational levels or through proteasome activity repression by proteasome inhibitors in order to reach an indirect p53 back-up level and potential activity (Soucy et al., 2009[138]; Zhu et al., 2013[177]). Recently, a polyphenolic alkanone, 6-Gingerol (6G), extracted from ginger (Zingiber officinale Roscoe) expressed anti-tumorigenic and pro-apoptotic activity against a variety of cancers. Thus, testing its efficacy in cervical cancer was promising. Findings showed its matching mechanism of action with BTZ to stimulate p53 expression along with its target p21. In addition to an increasing release of ubiquitinated proteins in 6G treated cells similar to that of the BTZ treatment. Interacting with proteosomal β-5 subunit was also seen but with a higher affinity than BTZ (Anderson et al., 2015[5]). In parallel, BTZ was able to perform its proteasomal activities in cervical cancer cells mediated by its tumor suppressors such as p53, hDlg and hScrib, especially when combined with cisplatin in another trial. Hence, valorizing the auspicious application of this dual therapy in resistant cervical cancer cases (Singha et al., 2015[135]). Based on this, BTZ efficacy in cervical cancer was marked yet a novel discovered medicine with better physiological characteristics limited its usage.

Colorectal cancer

Recently, it was proved that targeting poor proteasomal function with radioiodine will limit CT26 colon cancer stem cells resistance to BTZ therapy through multiple mechanisms in terms of sodium-iodide symporter (NIS) fusion protein that usually accumulates in cells with low proteasome activity. From one hand, CT26/NIS-cODC cells exhibited the assembly of NIS and took up of radioiodine under proteasome inhibitory conditions (Mehdizadeh et al., 2021[101]). From the other hand, CT26/NIS-cODC cells enriched for stemness cumulated NIS and executed a high radioiodine uptake. Also, CT26/NIS-cODC cell populations were able to survive following BTZ treatment because of their enrichment with CSCs. Furthermore, an administered 131I treatment strengthened the therapeutic efficacy of BTZ and suppressed cancer stemness along with a high radioiodine uptake in vivo. So, 131I therapy was seen to boost the efficacy of BTZ treatment against CT26/NIS-cODC tumors. Concluding that cancer stemness in vivo is increased by BTZ alone but is suppressed by adding 131I therapy (Rosenberg et al., 2008[130]). Dual BTZ and epirubicin treatment intensified tumor immunogenicity and the induction of antitumor immunity also. This therapeutic fusion potentiated immunogenic cell death induction of colorectal cancer cells by increasing expression of death receptors such as Fas, which enhanced colorectal cancer cells susceptibility to Fas/FasL mediated tumor cell killing but not healthy non-malignant epithelial cells, to apoptosis. Also, this regimen stabilized the transcriptional activation of Fas in colorectal cancer cells but not in normal cells (Gomez-Abuin et al., 2007[47]).

Endometrial cancer

Proteasome inhibitor (PIs) are well known to act on ubiquitin pathways. Recently, an underlying enzyme named as Ubiquitin C-terminal hydrolase L5 (UCHL5) was suggested to be a contributing factor for tumor growth progression and metastasis occurrence in multiple cancer tumors as endometrial cancer (EC). To identify the role of UCHL5 on EC, bioinformatics analysis revealed that UCHL5 overexpression in EC tissues lead to lower overall survival (Merrill et al., 2020[105]). Hence, UCHL5 was seen to be reversibly associated with the 26S proteasome in order to prevent target proteins degradation by hydrolyzing ubiquitin chains (Rastogi and Mishra, 2012[126]). This calls for further investigation of PI therapies that directly target enzymatic signaling pathways including UCHL5. Consequently Pis, mainly BTZ, were administered along with a panel of 23 chemotherapeutic agents and targeted drugs to organoid cultures of seven patients with different histological subtypes of endometrial cancer, including early-stage/grade endometrioid adenocarcinoma and high-grade serous carcinoma. The highest responsiveness was correlated to the combination of a histone deacetylase (HDAC) inhibitor and a proteasome inhibitor. Functionally, HDAC inhibitors and proteasome inhibitors were seen to express a dual proteasome and aggresome blockage resulting in apoptosis due to the accumulation of misfolded proteins (Bruning et al., 2011[19]). In another recent publication, BTZ combined to Histone Deacetylase Inhibitors were able to alleviate the impact of gain-of-function of p53 mutations through an enhanced p53 mutated cancer cell sensitivity towards BTZ caused by the preliminary addition of HDACi (Rastogi et al., 2015[125]). Obtained findings will serve as a crucial foundation for an improved combination therapy to be tested in vitro and in vivo reaching the main EC endpoints defined through the induction of apoptosis and the endometrial tumor regression.

Kidney cancer

In renal carcinoma, the proteasome inhibitor carfilzomib was studied and showed a significant acute and long-term cytotoxicity yet an in vivo anti-tumoral activity correlated with the level of accumulation of ubiquitinated proteins (UPs) (Chakraborty et al., 2012[23]). Also, researches tried to evaluate the prognostic role of the ubiquitin proteasome system (UPS) in renal tumors based on the categorization of involved genes. In fact, among 91 differentially expressed UPSs, 48 prognosis related genes were highlighted. These genes can be either subject to downregulation as ASB15, HECW1, RNF150, USP44, FBXO2 or upregulation as MDM4, KCTD13, TRAF2, PCGF1, TRAF3IP2. So, this destabilization of genetic expression marks UPS variations in renal carcinoma (Miyamoto et al., 2013[108]). Such findings impose the need for further studies on the abnormal ubiquitin proteasome system factors' as a main contributor in patient prognosis.

Nasopharyngeal cancer (NPC)

Nasopharyngeal carcinoma (NPC) reflects one of the complex tumor types since it is related to various environmental and host-dependent factors directly affecting cancerous progression. Majorly, Epstein Barr Virus (EBV) plays a key role in tissue growth, distant metastasis, and immune disability leading to a poor patient prognosis. Therefore, multiple intrinsic factors as EBV related onco-viral proteins such as Latent Membrane Protein family (LMP1, LMP2), Epstein Barr Nuclear Antigen 1 (EBNA1) and EBV-related glycoprotein B (gB) are proved to destabilize signaling pathways reaching a fast metastatic severe presentation of NPC (Meng et al., 2018[104]). Hence, activation signaling of proteasomes seems to be a crucial part of this overall critical mechanism. In vitro and in vivo investigations implemented the use of proteasome inhibitors in the therapeutic setting of NPC. Recently, Hui et al. tested the efficacy of BTZ combined to SAHA (Vorinostat) in female BALB/c nude (nu/nu) mice. Majorly, various apoptotic mechanisms and effects on lytic cycle activation of EBV were highlighted. This dual therapy synergistically induced killing of a panel of NPC cell lines and suppressed the growth of NPC xenografts in nude mice. Plus, BTZ limited SAHA's induction of EBV replication and hindered the production of infectious viral particles in NPC cells (Li et al., 2021[85]). In another study, Jiang et al. (2017[66]) studied the mechanism by which immune evasion affects the response to treatment of NPC. BTZ downregulated IFNγ-induced IDO expression via inhibition of JAK/STAT1 signaling pathway and promoted IkB-α phosphorylation-ubiquitination to release NF-kB from IkB-α (Guo et al., 2021[51]).

Breast cancer

The primary treatment of breast cancer depends mainly on several chemotherapy combinations. However, unsuccessful treatments and poor prognostic outcomes are still being reported. This indicates the need to use targeted therapies such as proteasome inhibitors. By the start of 2000s, proteasome inhibitors were considered novel promising compounds in multiple hematological malignancies and solid tumors, based on the preliminary investigations seen in MM (Hamadneh et al., 2021[53]). However, following trials downgraded the incorporation of a proteasome inhibitor in solid tumors, hence minimizing the focus on proving this hypothesis (Keefe et al., 2013[72]). A recent study tested the application of PIs on some triple-negative breast cancer cell lines (TNBCs) which were able to survive after being exposed to standard doses of BTZ or carfilzomib which can potentially block the b5 peptidase of the proteasome (Meng et al., 2020[103]). While at higher (PIs) concentrations, cellular deterioration was achieved through the additional inhibition of b1 and b2, valorizing the necessity to act all sites of action to achieve a successful response. At this stage, CRISPR gene editing approaches were implemented to prove the complete inactivation of b1 and b2 re-sensitized cells to BTZ and carfilzomib, both in vitro and in vivo (Zhou et al., 2020[173]). In parallel, Radhakrishnan et al. (2010[123]) identified the source of the acquired resistance mechanism toward PIs where a transcription factor Nrf1 upregulated a set of proteasome subunit genes, hence increasing the proteasome content and its defensive action (Tapia-Laliena et al., 2019[146]). In fact, as cancer is known to be complex in nature, a partial response to therapy is not enough. Thus, Weyburne et al. (2017[159]) confirmed the limited ability of PIs to be used as individual therapy in solid tumors treatment as their potential to limit b2 activity is quite absent. So, PIs must be accompanied with a specific b2 inhibitor in order to obtain effective response (Martin et al., 2019[99]; Zhao et al., 2016[172]). From a medicinal chemistry perspective, structuration of a dual b2/b5 inhibitor is a challenging path. For that, other research groups attempted to block sites upstream of the proteasome, such as E1 and E3 enzymes as well as p97 resulting in an incomplete solid tumor response (Hui et al., 2013[59]; Jiao et al., 2019[67]; Yarza et al., 2021[165]).

BTZ Use in Liquid TumorsMantle cell lymphoma (MCL)

Mantle cell lymphoma (MCL) is a rare, B cell non-Hodgkin's lymphoma with highly heterogeneous clinical presentation and aggressiveness (Jiang et al., 2017[66]). The inhibitory mechanism of proteasome exhibited MCL cell death through decreased NF-kappaB signaling and prevented IκB degradation and arrested cell cycle by limiting p27 breakdown while releasing high levels of reactive oxygen species (Fisher et al., 2006[39]; Hanel and Epperla, 2020[54]). BTZ was the first proteasome inhibitor to be FDA approved for relapsed/refractory MCL based on PINNACLE trial's findings (Goy et al., 2009[48]). Carfilzomib also showed promising effects in MCL with a lower encountered neurotoxicity compared to BTZ but the cardiotoxicity risk was more severe (Zhang et al., 2013[168]). Thus, BTZ is considered the unique proteasome inhibitor currently used with proven single-agent activity in relapsed/refractory MCL. Yet the efficacy of PIs in MCL cases is still based on their usage as combination therapies (Shah et al., 2018[134]). A brand new phase II randomized trial tested BTZ for its tolerability and efficacy either as a consolidation (BC) or maintenance (BM) therapy following immune-chemotherapy then autologous transplant (ASCT). Findings showed 54.1 % versus 64.4 % of 8-year PFS in BC and BM, respectively. Suggesting the promising value of BTZ in MCL practice (Lee et al., 2019[81]). While a phase II HOVON trial was based on administering BTZ as a maintenance therapy after R-CHOP, cytarabine and autologous stem cell transplantation in newly diagnosed patients with mantle cell lymphoma proved no added value for BTZ based on 63 % 5-year event free survival (EFS) versus 60 % 5-year (EFS) in BTZ and placebo group, respectively. Also, a 90 % 5 year overall survival (OS) was achieved in both groups (Kaplan et al., 2020[70]). These contradictory conclusions obtained impose the need for future improved hypothesis concerning BTZ use in MCL treatment regimens.

Leukemia

Acute lymphoblastic leukemia (ALL) is the most frequently diagnosed cancer in children (Doorduijn et al., 2020[37]). The combination of BTZ, rituximab, and a pediatric-inspired ALL regimen was seen to be active and well tolerated in de novo CD20+ Philadelphia-negative precursor B-ALL (Hunger and Mullighan, 2015[60]). In children with relapsed or refractory ALL, the addition of BTZ to re-induction chemotherapy that includes mitoxantrone exhibited a complete response in the majority of cases with minimal toxicity occurrence (August et al., 2020[6]). Also, children and young adults with Early T-Cell Precursor (ETP) Acute Lymphoblastic Leukemia suffered from poor prognosis compared with other T-ALL subtypes. At this stage, Venetoclax was used as a sole therapy until resistance cases emerged (Jain et al., 2021[63]). Venetoclax was initiated individually with successful preliminary antitumor activity reaching resistant cases (Bose et al., 2017[16]). In support of this hypothesis, recent research work demonstrated that BTZ can induce the BH3-only protein NOXA responsible to downregulate, among other Bcl-2 protein family members, myeloid cell leukemia-1 (Tahir et al., 2017[142]). In addition, BTZ successfully modulated the oncogenic T-ALL driver NOTCH1 by suppressing the expression of Notch and its target genes (HES1, GATA3, and RUNX3) in parallel to a synergistic activity with dexamethasone, leading to a near-complete remission of T-ALL xenografted tumors in vivo (Bassan et al., 2018[10]). These results strengthened the promising objectives of implementing a combination of less-toxic bioactive drugs for patients with highly resistant R/R T-ALL. Hence, venetoclax along with BTZ (VEBO) imposed an effective and well-tolerated chemotherapy-free strategy for R/R T-ALL, including ETP especially, as a bridge to transplantation in fit patients (Li et al., 2019[89]). In another clinical trial, BTZ utilization post-induction chemotherapy with the ALL REZ BFM 95/96 was associated with an immediate treatment efficacy significantly higher in patients treated with BTZ compared to the control group (85.7 % vs 57.6 %), respectively (Follini et al., 2019[40]). Assessment of BTZ efficacy in terms of improved survival in children with T-Cell Lymphoblastic Lymphoma (T-LL) was also studied. The endpoints of standard risk (SR) and intermediate risk (IR) patients with T-ALL and T-LL treated with BTZ were successfully achieved through an improved 3-year EFS and OS. Therapy intensification allowed elimination of cranial radiation (CXRT) in the majority of patients without excessive relapse (Batmanova et al., 2017[11]). Thus, proving the importance of BTZ in standard therapy for de novo T-LL appears advantageous. The implementation of BTZ in Plasma Cell Leukemia (PCL) as an induction therapy in combination with chemotherapy exhibited a 79 % overall response rate (ORR) (Jurczyszyn et al., 2018[69]; Teachey et al., 2020[147]). Its success was also maintained throughout Hematopoietic cell transplantation (HSCT) with an extended 14.3 months of median progression-free survival (PFS) and 15.7 months median overall survival (OS) (D'Arena et al., 2012[31]; Mahindra et al., 2012[94]; Royer et al., 2016[131]). Following transplantation, applying BTZ during maintenance phase showed its safe and feasible ability to lower risk of graft-versus host-disease (GvHD) as well as impairing the activation of T-cells and antigen-presenting cell (Alsina et al., 2014[4]; Jakubowiak et al., 2011[64]; Katodritou et al., 2018[71]; Kneppers et al., 2011[76]; Neben et al., 2012[115]; Nencioni et al., 2006[116]; Sonneveld et al., 2012[137]).

Promising Newly Discovered Pharmacological Effects of BTZ

BTZ therapeutic efficacy was not only limited to cancer tumors (Blanco et al., 2006[15]). In fact, BTZ was able to perform novel mechanisms of actions in terms of membranous nephropathy (MN), pulmonary hypertension (PH), neurofibromatosis and skin cancer (see Table 2(Tab. 2); References in Table 2: Chou and Talalay, 1984[24]; Cooper and Giancotti, 2014[29]; Fisher et al., 2006[39]; Geara et al., 2021[45]; Kim et al., 2012[74]; Lee et al., 2021[82]; Li et al., 2010[88], 2012[86], 2014[87]; Liu et al., 2021[90]; Meeth et al., 2016[100]; Nakagawa et al., 2000[113]; Synodos for NF2 Consortium et al., 2018[141]; Thompson, 2013[148]; Voltan, 2018[155]; Yalon et al., 2013[163]; Zhang et al., 2016[169]; Zhou and Hanemann, 2012[174]).

Conclusion

BTZ use is mainly approved for retreating patients with MM and as a first-line option for patients with mantle-cell lymphoma. Consequently, the prognostic impact of BTZ in patients with other liquid tumors and solid tumors is still under investigation. In our review, we detailed the conventional and advanced BTZ mechanisms of action which allowed BTZ to be a crucial option of MM anti-tumor regimen. Moreover, BTZ succeeded to exhibit its therapeutic potential in different types of solid and liquid tumors. Specifically, BTZ expressed better therapeutic efficacy in ovarian, endometrial and nasopharyngeal cancers. However, further investigations should be executed to adequately assess the promising value of BTZ in order to reach successful treatment outputs and better patient's prognosis. Furthermore, this review highlighted the newly discovered pharmacological effects of BTZ in multiple diseases such as membranous nephropathy, pulmonary hypertension and neurofibromatosis, which accredited BTZ with additional clinical value in cancer and other illnesses.

Notes

Mohammad Alwahsh and Joviana Farhat contributed equally as first author.

DeclarationAuthor contributions

Conceptualization, MA, JF, and RH; methodology, MA, JF; writing-review and editing, MA, JF, ST and LH; supervision, MA, LH and RH. All authors have read and agreed to the published version of the manuscript.

Funding

Not applicable.

Institutional review board statement

Not applicable.

Informed consent statement

Not applicable.

Acknowledgments

All authors acknowledge their respective institutions for their support.

Conflicts of interest

The authors declare no conflict of interest.

Adams J Discovery – velcade®: A new tool in the fight against multiple myeloma 2014 2022 June 07 National Cancer Institute Available from: https://www.cancer.gov/research/progress/discovery/velcade Adams J Proteasome inhibition in cancer: Development of PS-341 Semin Oncol 2001 28 6 613 619 10.1016/S0093-7754(01)90034-X Alsahafi E Begg K Amelio I Raulf N Lucarelli P Sauter T Clinical update on head and neck cancer: molecular biology and ongoing challenges Cell Death Dis 2019 10 8 540 10.1038/s41419-019-1769-9 Alsina M Becker PS Zhong X Adams A Hari P Rowley S Lenalidomide maintenance for high-risk multiple myeloma after allogeneic hematopoietic cell transplantation Biol Blood Marrow Transplant 2014 20 1183 1189 Anderson DJ Le Moigne R Djakovic S Kumar B Rice J Wong S Targeting the AAA ATPase p97 as an approach to treat cancer through disruption of protein homeostasis Cancer Cell 2015 28 653 665 10.1016/j.ccell.2015.10.002 August KJ Guest EM Lewing K Hays JA Gamis AS Treatment of children with relapsed and refractory acute lymphoblastic leukemia with mitoxantrone, vincristine, pegaspargase, dexamethasone, and bortezomib Pediatric Blood Cancer 2020 67 3 e28062 10.1002/pbc.28062 Bahlis NJ Sutherland H White D Sebag M Lentzsch S Kotb R Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma Blood 2018 132 2546 2554 10.1182/blood-2018-06-858852 Bard JAM Goodall EA Greene ER Jonsson E Dong KC Martin A Structure and function of the 26S proteasome Annu Rev Biochem 2018 87 697 724 10.1146/annurev-biochem-062917-011931 Bashraheel SS Domling A Goda SK Update on targeted cancer therapies, single or in combination, and their fine tuning for precision medicine Biomed Pharmacother 2020 125 110009 10.1016/j.biopha.2020.110009 Bassan R Bourquin JP DeAngelo DJ Chiaretti S New approaches to the management of adult acute lymphoblastic leukemia J Clin Oncol 2018 epub ahead of print 10.1200/jco.2017.77.3648 Batmanova N Shervashidze M Popa A Grivtsova L Serebryakova I Mentkevich G Bortezomib combination therapy of relapsed and refractory acute lymphoblastic leukemia in children Clin Oncohematol 2017 10 381–9 10.21320/2500-2139-2017-10-3-381-389 Becker SH Li H Heran Darwin K Biology and biochemistry of bacterial proteasomes Subcell Biochem 2019 93 339 358 10.1007/978-3-030-28151-9_11 Benvenuto M Ciuffa S Focaccetti C Sbardella D Fazi S Tundo GR The proteasome inhibitor bortezomib induces tongue, pharynx and salivary gland cancer cells death in vitro and delays tumor growth of salivary gland cancer cells transplanted in Mice PREPRINT (Version 1) available at Research Square 01 06 2021 10.21203/rs.3.rs-561856/v1 Bielskienė K Bagdonienė L Mozūraitienė J Kazbarienė B Janulionis E E3 ubiquitin ligases as drug targets and prognostic biomarkers in melanoma Medicina (Kaunas, Lithuania) 2015 51 1 1 9 10.1016/j.medici.2015.01.007 Blanco B Pérez-Simón JA Sánchez-Abarca LI Carvajal-Vergara X Mateos J Vidriales B Bortezomib induces selective depletion of alloreactive T lymphocytes and decreases the production of Th1 cytokines Blood 2006 107 3575 3583 10.1182/blood-2005-05-2118 Bose P Gandhi V Konopleva M Pathways and mechanisms of venetoclax resistance Leuk Lymphoma 2017 58 9 1 17 10.1080/10428194.2017.1283032 Boughton AJ Krueger S Fushman D Branching via K11 and K48 bestows ubiquitin chains with a unique interdomain interface and enhanced affinity for proteasomal subunit Rpn1 Structure 2020 28 1 29 43 10.1016/j.str.2019.10.008 Bray F Ferlay J Soerjomataram I Siegel RL Torre LA Jemal A Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries CA Cancer J Clin 2018 68 394 424 10.3322/caac.21492 Bruning A Vogel M Mylonas I Friese K Burges A Bortezomib targets the caspase-like proteasome activity in cervical cancer cells, triggering apoptosis that can be enhanced by nelfinavir Curr Cancer Drug Targets 2011 11 799 809 10.2174/156800911796798913 Carbone M Arron ST Beutler B Bononi A Cavenee W Cleaver JE Tumour predisposition and cancer syndromes as models to study gene-environment interactions Nat Rev Cancer 2020 20 533 549 10.1038/s41568-020-0265-y Cavo M Gay F Beksac M Pantani L Petrucci MT Dimopoulos MA Autologous haematopoietic stem-cell transplantation versus bortezomib-melphalan-prednisone, with or without bortezomib-lenalidomide-dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): a multicentre, randomised, open-label, phase 3 study Lancet Haematol 2020 7 6 e456 ee68 10.1016/s2352-3026(20)30099-5 Cerruti F Jocollè G Salio C Oliva L Paglietti L Alessandria B Proteasome stress sensitizes malignant pleural mesothelioma cells to bortezomib-induced apoptosis Sci Rep 2017 7 1 17626 10.1038/s41598-017-17977-9 Chakraborty D Bishayee K Ghosh S Biswas R Mandal SK Khuda-Bukhsh AR [6]-Gingerol induces caspase 3 dependent apoptosis and autophagy in cancer cells: drug-DNA interaction and expression of certain signal genes in HeLa cells Eur J Pharmacol 2012 694 20 29 10.1016/j.ejphar.2012.08.001 Chou T-C Talalay P Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors Adv Enzyme Regul 1984 22 27 55 Choueiri TK Tomczak P Park SH Venugopal B Ferguson T Chang Y-H Pembrolizumab versus placebo as post-nephrectomy adjuvant therapy for patients with renal cell carcinoma: Randomized, double-blind, phase III KEYNOTE-564 study J Clin Oncol 2021 39 18_Suppl LBA5 LLBA 10.1200/JCO.2021.39.15_suppl.LBA5 Chow LQM Head and neck cancer N Engl J Med 2020 382 60 72 10.1056/NEJMra1715715 Clague MJ Heride C Urbé S The demographics of the ubiquitin system Trends Cell Biol 2015 25 417 426 10.1016/j.tcb.2015.03.002 Collins GA Goldberg AL The logic of the 26S proteasome Cell 2017 169 792 806 10.1016/j.cell.2017.04.023 Cooper J Giancotti FG Molecular insights into NF2/Merlin tumor suppressor function FEBS Lett 2014 588 2743 2752 Dai X Bazaka K Thompson EW Ostrikov KK Cold atmospheric plasma: a promising controller of cancer cell states Cancers (Basel) 2020 12 11 3360 10.3390/cancers12113360 D'Arena G Valentini CG Pietrantuono G Guariglia R Martorelli MC Mansueto G Frontline chemotherapy with bortezomib-containing combinations improves response rate and survival in primary plasma cell leukemia: a retrospective study from GIMEMA Multiple Myeloma Working Party Ann Oncol 2012 23 1499 1502 10.1093/annonc/mdr480 Davis C Spaller BL Matouschek A Mechanisms of substrate recognition by the 26S proteasome Curr Opin Struct Biol 2021 67 161 169 10.1016/j.sbi.2020.10.010 de la Peña AH Goodall EA Gates SN Lander GC Martin A Substrate-engaged 26S proteasome structures reveal mechanisms for ATP-hydrolysis-driven translocation Science 2018 362 6418 eaav0725 10.1126/science.aav0725 Ding Z Fu Z Xu C Wang Y Wang Y Li J High-resolution cryo-EM structure of the proteasome in complex with ADP-AlFx Cell Res 2017 27 373 385 10.1038/cr.2017.12 Ding Z Xu C Sahu I Wang Y Fu Z Huang M Structural snapshots of 26S proteasome reveal tetraubiquitin-induced conformations Mol Cell 2019 73 1150 1161 10.1016/j.molcel.2019.01.018 Dong Y Zhang S Wu Z Li X Wang WL Zhu Y Cryo-EM structures and dynamics of substrate-engaged human 26S proteasome Nature 2019 565 7737 49 55 10.1038/s41586-018-0736-4 Doorduijn JK Zijlstra JM Lugtenburg PJ Kersten MJ Böhmer LH Minnema MC Bortezomib maintenance after R-CHOP, cytarabine and autologous stem cell transplantation in newly diagnosed patients with mantle cell lymphoma, results of a randomised phase II HOVON trial Brit J Haematol 2020 190 385 393 10.1111/bjh.16567 Durie BGM Hoering A Abidi MH Rajkumar SV Epstein J Kahanic SP Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial Lancet 2017 389 10068 519 527 10.1016/s0140-6736(16)31594-x Fisher RI Bernstein SH Kahl BS Djulbegovic B Robertson MJ de Vos S Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma J Clin Oncol 2006 24 4867 4874 10.1200/jco.2006.07.9665 Follini E Marchesini M Roti G Strategies to overcome resistance mechanisms in T-cell acute lymphoblastic leukemia Int J Mol Sci 2019 20 12 3021 10.3390/ijms20123021 Franchi M Vener C Garau D Kirchmayer U Di Martino M Romero M Bortezomib-based therapy in non-transplant multiple myeloma patients: a retrospective cohort study from the FABIO project Ther Adv Hematol 2021 12 2040620721996488 10.1177/2040620721996488 Fritze J Zhang M Luo Q Lu X An overview of the bacterial SsrA system modulating intracellular protein levels and activities Appl Microbiol Biotechnol 2020 104 5229 5241 10.1007/s00253-020-10623-x Galluzzi L Buqué A Kepp O Zitvogel L Kroemer G Immunogenic cell death in cancer and infectious disease Nat Rev Immunol 2017 17 97 111 10.1038/nri.2016.107 Galluzzi L Humeau J Buqué A Zitvogel L Kroemer G Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors Nat Rev Clin Oncol 2020 17 725 741 10.1038/s41571-020-0413-z Geara AS Bhoj V Hogan JJ Bortezomib treatment for refractory PLA2R-positive membranous nephropathy Glomerular Dis 2021 1 1 40 43 Gold LI Eggleton P Sweetwyne MT Van Duyn LB Greives MR Naylor SM Calreticulin: non-endoplasmic reticulum functions in physiology and disease FASEB J 2010 24 665 683 10.1096/fj.09-145482 Gomez-Abuin G Winquist E Stadler WM Pond G Degendorfer P Wright J A phase II study of PS-341 (Bortezomib) in advanced or metastatic urothelial cancer. A trial of the Princess Margaret Hospital and University of Chicago phase II consortia Invest New Drugs 2007 25 181 185 10.1007/s10637-006-9009-4 Goy A Bernstein SH Kahl BS Djulbegovic B Robertson MJ de Vos S Bortezomib in patients with relapsed or refractory mantle cell lymphoma: updated time-to-event analyses of the multicenter phase 2 PINNACLE study Ann Oncol 2009 20 520 525 10.1093/annonc/mdn656 Grosicki S Simonova M Spicka I Pour L Kriachok I Gavriatopoulou M Once-per-week selinexor, bortezomib, and dexamethasone versus twice-per-week bortezomib and dexamethasone in patients with multiple myeloma (BOSTON): a randomised, open-label, phase 3 trial Lancet 2020 396 10262 1563 1573 10.1016/s0140-6736(20)32292-3 Gulla A Morelli E Samur MK Botta C Hideshima T Bianchi G Bortezomib induces anti-multiple myeloma immune response mediated by cGAS/STING pathway activation Blood Cancer Discov 2021 2 468 483 10.1158/2643-3230.Bcd-21-0047 Guo H Li Y Liu Y Chen L Gao Z Zhang L Prognostic role of the ubiquitin proteasome system in clear cell renal cell carcinoma: a bioinformatic perspective J Cancer 2021 12 4134 4147 10.7150/jca.53760 Hallberg B Palmer RH Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology Nat Rev Cancer 2013 13 685 700 10.1038/nrc3580 Hamadneh L Abu-Irmaileh B Al-Majawleh M Bustanji Y Jarrar Y Al-Qirim T Doxorubicin-paclitaxel sequential treatment: insights of DNA methylation and gene expression changes of luminal A and triple negative breast cancer cell lines Mol Cell Biochem 2021 476 3647 3654 10.1007/s11010-021-04191-5 Hanel W Epperla N Emerging therapies in mantle cell lymphoma J Hematol Oncol 2020 13 1 79 10.1186/s13045-020-00914-1 Harsha C Banik K Ang HL Girisa S Vikkurthi R Parama D Targeting AKT/mTOR in oral cancer: mechanisms and advances in clinical trials Int J Mol Sci 2020 21 9 3285 10.3390/ijms21093285 Hopfner K-P Hornung V Molecular mechanisms and cellular functions of cGAS–STING signalling Nat Rev Mol Cell Biol 2020 21 501 521 10.1038/s41580-020-0244-x Hou H Sun D Zhang X The role of MDM2 amplification and overexpression in therapeutic resistance of malignant tumors Cancer Cell Int 2019 19 216 10.1186/s12935-019-0937-4 Huang Z Wu Y Zhou X Xu J Zhu W Shu Y Efficacy of therapy with bortezomib in solid tumors: a review based on 32 clinical trials Fut Oncol 2014 10 1795 1807 10.2217/fon.14.30 Hui KF Lam BH Ho DN Tsao SW Chiang AK Bortezomib and SAHA synergistically induce ROS-driven caspase-dependent apoptosis of nasopharyngeal carcinoma and block replication of Epstein-Barr virus Mol Cancer Ther 2013 12 747 758 10.1158/1535-7163.Mct-12-0811 Hunger SP Mullighan CG Acute lymphoblastic leukemia in children N Engl J Med 2015 373 1541 1552 10.1056/NEJMra1400972 Ibarra G Peña M Abril L Senín A Maluquer C Clapés V Dose intensity and treatment duration of bortezomib in transplant-ineligible newly diagnosed multiple myeloma Eur J Haematol 2021 107 246 254 10.1111/ejh.13643 Ibata S Sato T Kuroda H Nagamachi Y Iyama S Fujimi A A phase II trial of small-dose bortezomib, lenalidomide and dexamethasone (sVRD) as consolidation/maintenance therapy in patients with multiple myeloma Cancer Chemother Pharmacol 2016 78 1041 1049 10.1007/s00280-016-3163-y Jain H Sengar M Goli VB Thorat J Tembhare P Shetty D Bortezomib and rituximab in de novo adolescent/adult CD20-positive, Ph-negative pre-B-cell acute lymphoblastic leukemia Blood Adv 2021 5 3436 3444 10.1182/bloodadvances.2020003368 Jakubowiak AJ Griffith KA Reece DE Hofmeister CC Lonial S Zimmerman TM Lenalidomide, bortezomib, pegylated liposomal doxorubicin, and dexamethasone in newly diagnosed multiple myeloma: a phase 1/2 Multiple Myeloma Research Consortium trial Blood 2011 118 535 543 10.1182/blood-2011-02-334755 Jakubowiak AJ Kendall T Al-Zoubi A Khaled Y Mineishi S Ahmed A Phase II trial of combination therapy with bortezomib, pegylated liposomal doxorubicin, and dexamethasone in patients with newly diagnosed myeloma J Clin Oncol 2009 27 5015 5022 10.1200/jco.2008.19.5370 Jiang GM Wang HS Du J Ma WF Wang H Qiu Y Bortezomib relieves immune tolerance in nasopharyngeal carcinoma via STAT1 suppression and indoleamine 2,3-dioxygenase downregulation Cancer Immunol Res 2017 5 42 51 10.1158/2326-6066.Cir-16-0102 Jiao D Huan Y Zheng J Wei M Zheng G Han D UHRF1 promotes renal cell carcinoma progression through epigenetic regulation of TXNIP Oncogene 2019 38 5686 5699 10.1038/s41388-019-0822-6 Joshi NP Broughman JR Postoperative management of salivary gland tumors Curr Treat Options Oncol 2021 22 3 23 10.1007/s11864-021-00820-9 Jurczyszyn A Radocha J Davila J Fiala MA Gozzetti A Grząśko N Prognostic indicators in primary plasma cell leukaemia: a multicentre retrospective study of 117 patients Brit J Haematol 2018 180 831 839 10.1111/bjh.15092 Kaplan LD Maurer MJ Stock W Bartlett NL Fulton N Pettinger A Bortezomib consolidation or maintenance following immunochemotherapy and autologous stem cell transplantation for mantle cell lymphoma: CALGB/Alliance 50403 Am J Hematol 2020 95 583 593 10.1002/ajh.25783 Katodritou E Terpos E Delimpasi S Kotsopoulou M Michalis E Vadikolia C Real-world data on prognosis and outcome of primary plasma cell leukemia in the era of novel agents: a multicenter national study by the Greek Myeloma Study Group Blood Cancer J 2018 8 3 31 10.1038/s41408-018-0059-6 Keefe SM Nathanson KL Rathmell WK The molecular biology of renal cell carcinoma Semin Oncol 2013 40 421 428 10.1053/j.seminoncol.2013.05.006 Keruakous AR Day S Yuen C Disease biology alters the response to frontline bortezomib, lenalidomide and dexamethasone in Multiple Myeloma Glob J Cancer Ther 2021 7 1 010 015 10.17352/2581-5407.000037 Kim S-Y Lee J-H Huh JW Kim HJ Park MK Ro JY Bortezomib alleviates experimental pulmonary arterial hypertension Am J Respir Cell Mol Biol 2012 47 698 708 10.1165/rcmb.2011-0331OC Kitamura N Sento S Yoshizawa Y Sasabe E Kudo Y Yamamoto T Current trends and future prospects of molecular targeted therapy in head and neck squamous cell carcinoma Int J Mol Sci 2020 22 1 240 10.3390/ijms22010240 Kneppers E van der Holt B Kersten M-J Zweegman S Meijer E Huls G Lenalidomide maintenance after nonmyeloablative allogeneic stem cell transplantation in multiple myeloma is not feasible: results of the HOVON 76 Trial Blood 2011 118 2413 2419 10.1182/blood-2011-04-348292 Korde N Roschewski M Zingone A Kwok M Manasanch EE Bhutani M Treatment with carfilzomib-lenalidomide-dexamethasone with lenalidomide extension in patients with smoldering or newly diagnosed multiple myeloma JAMA Oncol 2015 1 746 754 10.1001/jamaoncol.2015.2010 Kroemer G Zitvogel L Subversion of calreticulin exposure as a strategy of immune escape Cancer Cell 2021 39 449 451 10.1016/j.ccell.2021.01.014 Kuku I Bayraktar MR Kaya E Erkurt MA Bayraktar N Cikim K Serum proinflammatory mediators at different periods of therapy in patients with multiple myeloma Mediators Inflamm 2005 3 171 174 10.1155/mi.2005.171 Laszlo V Valko Z Ozsvar J Kovacs I Garay T Hoda MA The FAK inhibitor BI 853520 inhibits spheroid formation and orthotopic tumor growth in malignant pleural mesothelioma J Mol Med 2019 97 231 242 10.1007/s00109-018-1725-7 Lee HJ Badillo M Romaguera J Wang M A phase II study of carfilzomib in the treatment of relapsed/refractory mantle cell lymphoma Brit J Haematol 2019 184 460 462 10.1111/bjh.15107 Lee MS Lim SH Yu AR Hwang CY Kang I Yeo EJ Carfilzomib in combination with bortezomib enhances apoptotic cell death in B16-F1 melanoma cells Biology 2021 10 2 153 10.3390/biology10020153 Lee YT Tan YJ Oon CE Molecular targeted therapy: Treating cancer with specificity Eur J Pharmacol 2018 834 188 196 10.1016/j.ejphar.2018.07.034 Li C Zhang Y Cheng X Yuan H Zhu S Liu J PINK1 and PARK2 suppress pancreatic tumorigenesis through control of mitochondrial iron-mediated immunometabolism Dev Cell 2018 46 441 455 10.1016/j.devcel.2018.07.012 Li J Pohl L Schüler J Korzeniewski N Reimold P Kaczorowski A Targeting the proteasome in advanced renal cell carcinoma: complexity and limitations of patient-individualized preclinical drug discovery Biomedicines 2021 9 6 627 10.3390/biomedicines9060627 Li W Cooper J Karajannis MA Giancotti FG Merlin: a tumour suppressor with functions at the cell cortex and in the nucleus EMBO Rep 2012 13 204 215 10.1038/embor.2012.11 Li W Cooper J Zhou L Yang C Erdjument-Bromage H Zagzag D Merlin/NF2 loss-driven tumorigenesis linked to CRL4DCAF1-mediated inhibition of the Hippo pathway kinases Lats1 and 2 in the nucleus Cancer Cell 2014 26 1 48 60 10.1016/j.ccr.2014.05.001 Li W You L Cooper J Schiavon G Pepe-Caprio A Zhou L Merlin/NF2 suppresses tumorigenesis by inhibiting the E3 ubiquitin ligase CRL4DCAF1 in the nucleus Cell 2010 140 4 477 490 10.1016/j.cell.2010.01.029 Li Z He S Look AT The MCL1-specific inhibitor S63845 acts synergistically with venetoclax/ABT-199 to induce apoptosis in T-cell acute lymphoblastic leukemia cells Leukemia 2019 33 262 266 10.1038/s41375-018-0201-2 Liu Z Zeng Q Xiang B Bortezomib-based regimens improve the prognosis of newly diagnosed MM patients with chromosomal aberrations except for del (17q13): A retrospective study from a single center Medicine (Baltimore) 2021 100 18 e25834 10.1097/MD.0000000000025834 Lo Nigro C Denaro N Merlotti A Merlano M Head and neck cancer: improving outcomes with a multidisciplinary approach Cancer Manag Res 2017 9 363 371 10.2147/cmar.S115761 Longworth J Dittmar G Assessment of ubiquitin chain topology by targeted mass spectrometry Methods Mol Biol 2019 1977 25 34 10.1007/978-1-4939-9232-4_3 Łuczkowska K Sokolowska KE Taryma-Lesniak O Pastuszak K Supernat A Bybjerg-Grauholm J Bortezomib induces methylation changes in neuroblastoma cells that appear to play a significant role in resistance development to this compound Sci Rep 2021 11 1 9846 10.1038/s41598-021-89128-0 Mahindra A Kalaycio ME Vela-Ojeda J Vesole DH Zhang M-J Li P Hematopoietic cell transplantation for primary plasma cell leukemia: results from the Center for International Blood and Marrow Transplant Research Leukemia 2012 26 1091 1097 10.1038/leu.2011.312 Majumder P Baumeister W Proteasomes: unfoldase-assisted protein degradation machines Biol Chem 2019 401 183 199 10.1515/hsz-2019-0344 Majumder P Rudack T Beck F Danev R Pfeifer G Nagy I Cryo-EM structures of the archaeal PAN-proteasome reveal an around-the-ring ATPase cycle Proc Natl Acad Sci U S A 2019 116 534 539 10.1073/pnas.1817752116 Malcovati L Rumi E Cazzola M Somatic mutations of calreticulin in myeloproliferative neoplasms and myelodysplastic/myeloproliferative neoplasms Haematologica 2014 99 1650 1652 10.3324/haematol.2014.113944 Mao Y Structure, dynamics and function of the 26S proteasome Subcellular Biochem 2021 96 1 151 10.1007/978-3-030-58971-4_1 Martin P Ruan J Furman R Rutherford S Allan J Chen Z A phase I trial of palbociclib plus bortezomib in previously treated mantle cell lymphoma Leuk Lymphoma 2019 60 2917 2921 10.1080/10428194.2019.1612062 Meeth K Wang JX Micevic G Damsky W Bosenberg MW The YUMM lines: a series of congenic mouse melanoma cell lines with defined genetic alterations Pigment Cell Melanoma Res 2016 29 590 597 10.1111/pcmr.12498 Mehdizadeh K Ataei F Hosseinkhani S Treating MCF7 breast cancer cell with proteasome inhibitor Bortezomib restores apoptotic factors and sensitizes cell to Docetaxel Med Oncol 2021 38 6 64 10.1007/s12032-021-01509-7 Mendes ML Fougeras MR Dittmar G Analysis of ubiquitin signaling and chain topology cross-talk Journal of proteomics 2020 215 103634 10.1016/j.jprot.2020.103634 Meng X Xiong Z Xiao W Yuan C Wang C Huang Y Downregulation of ubiquitin-specific protease 2 possesses prognostic and diagnostic value and promotes the clear cell renal cell carcinoma progression Ann Transl Med 2020 8 6 319 10.21037/atm.2020.02.141 Meng X Yang S Li Y Li Y Devor EJ Bi J Combination of proteasome and histone deacetylase inhibitors overcomes the impact of gain-of-function p53 mutations Dis Markers 2018 2018 3810108 10.1155/2018/3810108 Merrill NM Vandecan NM Day KC Palmbos PL Day ML Udager AM MEK is a promising target in the basal subtype of bladder cancer Oncotarget 2020 11 3921 3932 10.18632/oncotarget.27767 Michalak M Groenendyk J Szabo E Gold LI Opas M Calreticulin, a multi-process calcium-buffering chaperone of the endoplasmic reticulum Biochem J 2009 417 651 666 10.1042/bj20081847 Mirabella AC Pletnev AA Downey SL Florea BI Shabaneh TB Britton M Specific cell-permeable inhibitor of proteasome trypsin-like sites selectively sensitizes myeloma cells to bortezomib and carfilzomib Chem Biol 2011 18 608 618 10.1016/j.chembiol.2011.02.015 Miyamoto Y Nakagawa S Wada-Hiraike O Seiki T Tanikawa M Hiraike H Sequential effects of the proteasome inhibitor bortezomib and chemotherapeutic agents in uterine cervical cancer cell lines Oncol Rep 2013 29 51 57 10.3892/or.2012.2072 Montagnani F Fornaro L Frumento P Vivaldi C Falcone A Fioretto L Multimodality treatment of locally advanced squamous cell carcinoma of the oesophagus: A comprehensive review and network meta-analysis Crit Rev Oncol Hematol 2017 114 24 32 10.1016/j.critrevonc.2017.03.024 Montes de Oca R Alavi AS Vitali N Bhattacharya S Blackwell C Patel K Belantamab mafodotin (GSK2857916) drives immunogenic cell death and immune-mediated antitumor responses in vivo Mol Cancer Ther 2021 20 1941 1955 10.1158/1535-7163.Mct-21-0035 Muenchow A Weller S Hinterleitner C Malenke E Bugl S Wirths S The BCL-2 selective inhibitor ABT-199 sensitizes soft tissue sarcomas to proteasome inhibition by a concerted mechanism requiring BAX and NOXA Cell Death Dis 2020 11 8 701 10.1038/s41419-020-02910-2 Nadhan R Srinivas P Pillai MR RTKs in pathobiology of head and neck cancers Adv Cancer Res 2020 147 319 373 10.1016/bs.acr.2020.04.008 Nakagawa T Zhu H Morishima N Li E Xu J Yankner BA Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-β Nature 2000 403 6765 98 103 10.1038/47513 Nangalia J Massie CE Baxter EJ Nice FL Gundem G Wedge DC Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2 N Engl J Med 2013 369 2391 2405 10.1056/NEJMoa1312542 Neben K Lokhorst HM Jauch A Bertsch U Hielscher T van der Holt B Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p Blood 2012 119 940 948 10.1182/blood-2011-09-379164 Nencioni A Schwarzenberg K Brauer KM Schmidt SM Ballestrero A Grunebach F Proteasome inhibitor bortezomib modulates TLR4-induced dendritic cell activation Blood 2006 108 551 558 10.1182/blood-2005-08-3494 Niu M Yi M Li N Wu K Wu K Advances of targeted therapy for hepatocellular carcinoma Front Oncol 2021 11 719896 10.3389/fonc.2021.719896 O'Donnell EK Laubach JP Yee AJ Chen T Huff CA Basile FG A phase 2 study of modified lenalidomide, bortezomib and dexamethasone in transplant-ineligible multiple myeloma Brit J Haematol 2018 182 222 230 10.1111/bjh.15261 Oh E Akopian D Rape M Principles of ubiquitin-dependent signaling Annu Rev Cell Dev Biol 2018 34 137 162 10.1146/annurev-cellbio-100617-062802 Okazuka K Ishida T Nashimoto J Uto Y Sato K Miyazaki K The efficacy and safety of modified bortezomib-lenalidomide-dexamethasone in transplant-eligible patients with newly diagnosed multiple myeloma Eur J Haematol 2020 104 110 115 10.1111/ejh.13349 Parsons S Maldonado EB Prasad V Comparison of drugs used for adjuvant and metastatic therapy of colon, breast, and non-small cell lung cancers JAMA Netw Open 2020 3 4 e202488 10.1001/jamanetworkopen.2020.2488 Pettersson S Sczaniecka M McLaren L Russell F Gladstone K Hupp T Non-degradative ubiquitination of the Notch1 receptor by the E3 ligase MDM2 activates the Notch signalling pathway Biochem J 2013 450 523 536 10.1042/bj20121249 Radhakrishnan SK Lee CS Young P Beskow A Chan JY Deshaies RJ Transcription factor Nrf1 mediates the proteasome recovery pathway after proteasome inhibition in mammalian cells Mol Cell 2010 38 17 28 10.1016/j.molcel.2010.02.029 Rajkumar SV Myeloma today: Disease definitions and treatment advances Am J Hematol 2016 91 90 100 10.1002/ajh.24236 Rastogi N Duggal S Singh SK Porwal K Srivastava VK Maurya R Proteasome inhibition mediates p53 reactivation and anti-cancer activity of 6-gingerol in cervical cancer cells Oncotarget 2015 6 43310 43325 10.18632/oncotarget.6383 Rastogi N Mishra DP Therapeutic targeting of cancer cell cycle using proteasome inhibitors Cell Div 2012 7 1 26 10.1186/1747-1028-7-26 Raza MH Siraj S Arshad A Waheed U Aldakheel F Alduraywish S ROS-modulated therapeutic approaches in cancer treatment J Cancer Res Clin Oncol 2017 143 1789 1809 10.1007/s00432-017-2464-9 Renrick AN Thounaojam MC de Aquino MTP Chaudhuri E Pandhare J Dash C Bortezomib sustains T cell function by inducing miR-155-mediated downregulation of SOCS1 and SHIP1 Front Immunol 2021 12 607044 10.3389/fimmu.2021.607044 Richardson PG Baz R Wang M Jakubowiak AJ Laubach JP Harvey RD Phase 1 study of twice-weekly ixazomib, an oral proteasome inhibitor, in relapsed/refractory multiple myeloma patients Blood 2014 124 1038 1046 10.1182/blood-2014-01-548826 Rosenberg JE Halabi S Sanford BL Himelstein AL Atkins JN Hohl RJ Phase II study of bortezomib in patients with previously treated advanced urothelial tract transitional cell carcinoma: CALGB 90207 Ann Oncol 2008 19 946 950 10.1093/annonc/mdm600 Royer B Minvielle S Diouf M Roussel M Karlin L Hulin C Bortezomib, doxorubicin, cyclophosphamide, dexamethasone induction followed by stem cell transplantation for primary plasma cell leukemia: a prospective phase II study of the Intergroupe Francophone du Myélome J Clin Oncol 2016 34 18 2125 2132 10.1200/JCO.2015.63.1929 Sabbah DA Hajjo R Sweidan K Review on Epidermal Growth Factor Receptor (EGFR) structure, signaling pathways, interactions, and recent updates of EGFR inhibitors Curr Top Med Chem 2020 20 815 834 10.2174/1568026620666200303123102 Schweitzer A Aufderheide A Rudack T Beck F Pfeifer G Plitzko JM Structure of the human 26S proteasome at a resolution of 3.9 Å Proc Natl Acad Sci U S A 2016 113 7816 7821 10.1073/pnas.1608050113 Shah C Bishnoi R Jain A Bejjanki H Xiong S Wang Y Cardiotoxicity associated with carfilzomib: systematic review and meta-analysis Leuk Lymphoma 2018 59 2557 2569 10.1080/10428194.2018.1437269 Singha B Gatla HR Phyo S Patel A Chen ZS Vancurova I IKK inhibition increases bortezomib effectiveness in ovarian cancer Oncotarget 2015 6 26347 26358 10.18632/oncotarget.4713 Sistigu A Yamazaki T Vacchelli E Chaba K Enot DP Adam J Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy Nat Med 2014 20 1301 1309 10.1038/nm.3708 Sonneveld P Schmidt-Wolf IG van der Holt B El Jarari L Bertsch U Salwender H Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON-65/GMMG-HD4 trial J Clin Oncol 2012 30 2946 2955 10.1200/JCO.2011.39.6820 Soucy TA Smith PG Milhollen MA Berger AJ Gavin JM Adhikari S An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer Nature 2009 458 7239 732 736 10.1038/nature07884 Spisek R Charalambous A Mazumder A Vesole DH Jagannath S Dhodapkar MV Bortezomib enhances dendritic cell (DC)-mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications Blood 2007 109 4839 4845 10.1182/blood-2006-10-054221 Su Z Han S Jin Q Zhou N Lu J Shangguan F Ciclopirox and bortezomib synergistically inhibits glioblastoma multiforme growth via simultaneously enhancing JNK/p38 MAPK and NF-κB signaling Cell Death Dis 2021 12 3 251 10.1038/s41419-021-03535-9 Synodos for NF2 Consortium Allaway R, Angus SP, Beauchamp RL, Blakeley JO, Bott M, Burns SS, Traditional and systems biology based drug discovery for the rare tumor syndrome neurofibromatosis type 2 PLoS One 2018 13 6 e0197350 10.1371/journal.pone.0197350 Tahir SK Smith ML Hessler P Rapp LR Idler KB Park CH Potential mechanisms of resistance to venetoclax and strategies to circumvent it BMC Cancer 2017 17 1 399 10.1186/s12885-017-3383-5 Takács A Lajkó E Láng O Istenes I Kőhidai L Alpha-lipoic acid alters the antitumor effect of bortezomib in melanoma cells in vitro Sci Rep 2020 10 1 14287 10.1038/s41598-020-71138-z Tallarida RJ Quantitative methods for assessing drug synergism Genes Cancer 2011 2 1003 1008 10.1177/1947601912440575 Taniguchi K Karin M NF-κB, inflammation, immunity and cancer: coming of age Nat Rev Immunol 2018 18 309 324 10.1038/nri.2017.142 Tapia-Laliena Korzeniewski N Peña-Llopis S Scholl C Fröhling S Hohenfellner M Cullin 5 is a novel candidate tumor suppressor in renal cell carcinoma involved in the maintenance of genome stability Oncogenesis 2019 8 1 4 10.1038/s41389-018-0110-2 Teachey DT Devidas M Wood BL Chen Z Hayashi RJ Annett RD Cranial radiation can be eliminated in most children with T-Cell Acute Lymphoblastic Leukemia (T-ALL) and bortezomib potentially improves survival in children with T-Cell Lymphoblastic Lymphoma (T-LL): Results of Children's Oncology Group (COG) Trial AALL1231 Blood 2020 136 Suppl 1 11 12 10.1182/blood-2020-134730 Thompson JL Carfilzomib: a second-generation proteasome inhibitor for the treatment of relapsed and refractory multiple myeloma Ann Pharmacother 2013 47 56 62 10.1345/aph.1R561 Tsumagari K Abd Elmageed ZY Sholl AB Green EA Sobti S Khan AR Bortezomib sensitizes thyroid cancer to BRAF inhibitor in vitro and in vivo Endocr Relat Cancer 2018 25 1 99 109 10.1530/ERC-17-0182 Tutt A Garber JE Kaufman B Viale G Fumagalli D Rastogi P OlympiA: A phase III, multicenter, randomized, placebo-controlled trial of adjuvant olaparib after (neo)adjuvant chemotherapy in patients with germline BRCA1/2 mutations and high-risk HER2-negative early breast cancer J Clin Oncol 2021 39 18_Suppl LBA1 LLBA 10.1200/JCO.2021.39.15_suppl.LBA1 U.S. FDA, U.S. Food and Drug Administration FDA approves durvalumab after chemoradiation for unresectable stage III NSCLC 2018 2022 June 06 White Oak, MD U.S. FDA Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-durvalumab-after-chemoradiation-unresectable-stage-iii-nsclc U.S. FDA, U.S. Food and Drug Administration FDA approves osimertinib as adjuvant therapy for non-small cell lung 2020 2022 June 06 White Oak, MD U.S. FDA Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-osimertinib-adjuvant-therapy-non-small-cell-lung-cancer-egfr-mutations U.S. FDA, U.S. Food and Drug Administration Table of surrogate endpoints that were the basis of drug approval or licensure 2022 2022 June 06 White Oak, MD U.S. FDA Available from: https://www.fda.gov/drugs/development-resources/table-surrogate-endpoints-were-basis-drug-approval-or-licensure Vesole DH Bilotti E Richter JR McNeill A McBride L Raucci L Phase I study of carfilzomib, lenalidomide, vorinostat, and dexamethasone in patients with relapsed and/or refractory multiple myeloma Brit J Haematol 2015 171 1 52 59 10.1111/bjh.13517 Voltan R p53 and merlin tumor suppressors: Two of a kind EBioMedicine 2018 37 23 24 10.1016/j.ebiom.2018.10.062 Wakelee HA Altorki NK Zhou C Csőszi T Vynnychenko IO Goloborodko O IMpower010: Primary results of a phase III global study of atezolizumab versus best supportive care after adjuvant chemotherapy in resected stage IB-IIIA non-small cell lung cancer (NSCLC) J Clin Oncol 2021 39 15_Suppl 8500 10.1200/JCO.2021.39.15_suppl.8500 Wang X Cimermancic P Yu C Schweitzer A Chopra N Engel JL Molecular details underlying dynamic structures and regulation of the human 26S proteasome Mol Cell Proteomics 2017 16 840 854 10.1074/mcp.M116.065326 Wehmer M Rudack T Beck F Aufderheide A Pfeifer G Plitzko JM Structural insights into the functional cycle of the ATPase module of the 26S proteasome Proc Natl Acad Sci U S A 2017 114 1305 1310 10.1073/pnas.1621129114 Weyburne ES Wilkins OM Sha Z Williams DA Pletnev AA de Bruin G Inhibition of the proteasome β2 site sensitizes triple-negative breast cancer cells to β5 inhibitors and suppresses Nrf1 activation Cell Chem Biol 2017 24 218 230 10.1016/j.chembiol.2016.12.016 Worden EJ Dong KC Martin A An AAA motor-driven mechanical switch in Rpn11 controls deubiquitination at the 26S proteasome Mol Cell 2017 67 799 811 10.1016/j.molcel.2017.07.023 Wu Y-H Wu W-S Lin L-C Liu C-S Ho S-Y Wang B Jr Bortezomib enhances radiosensitivity in oral cancer through inducing autophagy-mediated TRAF6 oncoprotein degradation J Exp Clin Cancer Res 2018 37 1 91 10.1186/s13046-018-0760-0 Wustrow D Zhou H-J Rolfe M Chapter Fourteen - Inhibition of ubiquitin proteasome system enzymes for anticancer therapy Annu Rep Med Chem 2013 48 205 225 10.1016/B978-0-12-417150-3.00014-4 Yalon M Rood B MacDonald TJ McCowage G Kane R Constantini S A feasibility and efficacy study of rapamycin and erlotinib for recurrent pediatric low‐grade glioma (LGG) Pediatr Blood Cancer 2013 60 1 71 76 10.1002/pbc.24142 Yang J LeBlanc FR Dighe SA Hamele CE Olson TL Feith DJ TRAIL mediates and sustains constitutive NF-κB activation in LGL leukemia Blood 2018 131 2803 2815 10.1182/blood-2017-09-808816 Yarza R Bover M Agulló-Ortuño MT Iglesias-Docampo LC Current approach and novel perspectives in nasopharyngeal carcinoma: the role of targeting proteasome dysregulation as a molecular landmark in nasopharyngeal cancer J Exp Clin Cancer Res 2021 40 1 202 10.1186/s13046-021-02010-9 Yau NK Fong AY Leung HF Verhoeft KR Lim QY Lam WY A Pan-Cancer review of ALK mutations: implications for carcinogenesis and therapy Curr Cancer Drug Targets 2015 15 327 336 10.2174/1568009615666150225123712 Yau RG Doerner K Castellanos ER Haakonsen DL Werner A Wang N Assembly and function of heterotypic ubiquitin chains in cell-cycle and protein quality control Cell 2017 171 918 933 10.1016/j.cell.2017.09.040 Zhang L Pham LV Newberry KJ Ou Z Liang R Qian J In vitro and in vivo therapeutic efficacy of carfilzomib in mantle cell lymphoma: targeting the immunoproteasome Mol Cancer Ther 2013 12 2494 2504 10.1158/1535-7163.Mct-13-0156 Zhang N Fu J-N Chou T-C Synergistic combination of microtubule targeting anticancer fludelone with cytoprotective panaxytriol derived from panax ginseng against MX-1 cells in vitro: experimental design and data analysis using the combination index method Am J Cancer Res 2016 6 1 97 104 Zhang X Lin C Song J Chen H Chen X Ren L Parkin facilitates proteasome inhibitor-induced apoptosis via suppression of NF-κB activity in hepatocellular carcinoma Cell Death Dis 2019 10 10 719 10.1038/s41419-019-1881-x Zhang Y Yang B Zhao J Li X Zhang L Zhai Z Proteasome Inhibitor Carbobenzoxy-L-Leucyl-L-Leucyl-L-Leucinal (MG132) enhances therapeutic effect of paclitaxel on breast cancer by inhibiting nuclear factor (NF)-κB signaling Med Sci Monit 2018 24 294 304 10.12659/msm.908139 Zhao W Zhou J Deng Z Gao Y Cheng Y SPOP promotes tumor progression via activation of β-catenin/TCF4 complex in clear cell renal cell carcinoma Int J Oncol 2016 49 1001 1008 10.3892/ijo.2016.3609 Zhou J Wang T Qiu T Chen Z Ma X Zhang L Ubiquitin-specific protease-44 inhibits the proliferation and migration of cells via inhibition of JNK pathway in clear cell renal cell carcinoma BMC Cancer 2020 20 1 214 10.1186/s12885-020-6713-y Zhou L Hanemann CO Merlin, a multi-suppressor from cell membrane to the nucleus FEBS Lett 2012 586 1403 1408 10.1016/j.febslet.2012.03.016 Zhu Y An X Zhang X Qiao Y Zheng T Li X STING: a master regulator in the cancer-immunity cycle Mol Cancer 2019 18 1 152 10.1186/s12943-019-1087-y Zhu Y Wang WL Yu D Ouyang Q Lu Y Mao Y Structural mechanism for nucleotide-driven remodeling of the AAA-ATPase unfoldase in the activated human 26S proteasome Nat Commun 2018 9 1 1360 10.1038/s41467-018-03785-w Zhu YX Kortuem KM Stewart AK Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma Leuk Lymphoma 2013 54 683 687 10.3109/10428194.2012.728597 Zitvogel L Rusakiewicz S Routy B Ayyoub M Kroemer G Immunological off-target effects of imatinib Nat Rev Clin Oncol 2016 13 431 446 10.1038/nrclinonc.2016.41 BTZ efficacy in other solid tumors Novel BTZ application in multiple diseases