Next Article in Journal
Predicting Lung Function Using Biomarkers in Alpha-1 Antitrypsin Deficiency
Next Article in Special Issue
Biological Effects of Small Sized Graphene Oxide Nanosheets on Human Leukocytes
Previous Article in Journal
Unraveling Lifelong Brain Morphometric Dynamics: A Protocol for Systematic Review and Meta-Analysis in Healthy Neurodevelopment and Ageing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nab-Paclitaxel in the Treatment of Gastrointestinal Cancers—Improvements in Clinical Efficacy and Safety

by
Md Sazzad Hassan
1,2,*,†,
Niranjan Awasthi
1,2,†,
Saisantosh Ponna
3 and
Urs von Holzen
1,2,4,5
1
Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA
2
Harper Cancer Research Institute, South Bend, IN 46617, USA
3
Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN 46556, USA
4
Goshen Center for Cancer Care, Goshen, IN 46526, USA
5
Department of Surgery, University of Basel School of Medicine, 4001 Basel, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2023, 11(7), 2000; https://doi.org/10.3390/biomedicines11072000
Submission received: 17 May 2023 / Revised: 3 July 2023 / Accepted: 10 July 2023 / Published: 15 July 2023
(This article belongs to the Special Issue Nanotechnology to Counter the Off-Target Toxicity of Therapeutics)

Abstract

:
Taxanes (paclitaxel and docetaxel) are one of the most useful classes of anticancer drugs. Taxanes are highly hydrophobic; therefore, these drugs must be dissolved in organic solvents (polysorbate or Cremophor EL), which contribute to their toxicities. To reduce this toxicity and to enhance their efficacy, novel formulations have been developed. Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is an albumin-stabilized, Cremophor-free, and water-soluble nanoparticle formulation of paclitaxel. Nab-paclitaxel has better solubility and less infusion-associated toxicity compared to solvent-based paclitaxel. Additionally, nab-paclitaxel can be given at higher doses and concentrations compared with solvent-based paclitaxel. Based on its superior clinical efficacy and safety profile, nab-paclitaxel received FDA approval for metastatic breast cancer (2008) and NSCLC (2011). Among gastrointestinal cancers, it is now approved in the USA for treating patients with metastatic adenocarcinoma of the pancreas as first-line therapy in combination with gemcitabine. Furthermore, several clinical trials have suggested the potential efficacy of nab-paclitaxel as a single agent or in combination with other agents for the treatment of metastatic esophageal, gastric, bowel, and biliary tract cancers. Nab-paclitaxel has been demonstrated to have greater overall response rates (ORR) with enhanced progression-free survival (PFS), overall survival (OS) and a superior safety profile with fewer adverse effects in patients with gastrointestinal tract cancers. This review summarizes the advantages associated with nab-paclitaxel-based regimens in terms of improving clinical efficacy and the safety profile in upper gastrointestinal cancer.

1. Introduction

The taxanes paclitaxel and docetaxel are among the most important antineoplastic drugs that function as microtubule-stabilizing agents, leading to the inhibition of cell mitosis. Paclitaxel and docetaxel were first approved by the Food and Drug Administration (FDA) for clinical use in the United States (US) in 1994 and 2004, respectively [1,2]. These taxanes are extensively used to treat various cancers, including gastrointestinal cancers such as esophageal, gastric, and pancreatic cancers. Although these taxanes are clinically very effective in treating a variety of cancers, they have very serious adverse effects that often hinder their regular use. The solvents used to dissolve taxanes have a major contribution towards toxicities associated with taxanes [3,4]. Since taxanes are hydrophobic, Cremophor EL (CrEL) is used to dissolve paclitaxel, and polysorbate-80 is used to dissolve docetaxel [4]. These solvents contribute to considerable adverse effects such as hypersensitivity reactions, including anaphylactic reactions and neuropathies [3,4]. Additionally, these solvents impair tumor penetration, limiting the clinical effectiveness of paclitaxel and docetaxel [3,4]. Furthermore, the uptake of taxanes by cells is impaired by P-glycoprotein, which actively exports paclitaxel out of the cells, leading to paclitaxel resistance [5,6].
Due to the significant toxicities associated with the clinical use of solvent-based taxanes, numerous efforts have been made to overcome this issue. The development of a nanoparticle albumin-bound formulation of paclitaxel (nab-paclitaxel) is a result of these efforts. Nab-paclitaxel is a colloidal suspension of Cremophor solvent-free albumin-bound nanoparticle (mean diameter ~130 nm) of paclitaxel, which is homogenized with human serum albumin in an aqueous solution [7]. This water soluble nab-paclitaxel has appeared as the favored treatment option due to its superior clinical efficacy and lower incidence of toxicity compared to solvent-based paclitaxel [8]. The tolerable dose of nab-paclitaxel is much higher than paclitaxel with fewer side effects [7]. In a mouse xenograft model, it has been shown that the maximum tolerable dose (MTD) of nab-paclitaxel was much higher (30 mg/kg/day) than that of cremophor solvent-based paclitaxel (13 mg/kg/day) [9]. Similarly, in humans, the MTD of nab-paclitaxel was higher (300 mg/m2, every three weeks) compared to that of solvent-based paclitaxel (250 mg/m2, every three weeks) with significantly fewer adverse effects [10,11]. Nab-paclitaxel (Abraxane®) was approved by the FDA in the USA in January 2005 for use in patients with metastatic breast cancer and in October 2012 for use in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) [12,13]. Nab-paclitaxel, in combination with gemcitabine, is also approved in the USA for treating advanced pancreatic cancer [14]. The development of nab-paclitaxel became possible with the advent of the medical applications of drug nanotechnology. Drug nanotechnology involves the design and development of novel drugs that carry nanoparticles (Figure 1) [15]. The molecular structure of nab-paclitaxel, as depicted graphically in Figure 1, consists of a hydrophobic center containing paclitaxel due to its lack of solubility in water. The external hydrophilic shell is made of human serum albumin, which has negatively charged amino acids. The size of the molecule ranges from 50 to 150 nm, with a mean diameter of around 130 nm. The negatively charged amino acids of albumin repel each other, keeping the molecules in a homogenous aqueous suspension. Nab-paclitaxel essentially functions similar to solvent-based paclitaxel and other taxanes by binding to a pocket in the β-tubulin, thereby disrupting microtubule organization leading to the suppression of cell mitosis, as shown graphically in Figure 2. With this new albumin-bound formulation, nab-paclitaxel can cross endothelial cells by binding with an albumin receptor known as gp60, which is expressed on the surface of the endothelium, as shown graphically in Figure 3 [16]. Thus, unlike solvent-based paclitaxel, nab-paclitaxel exploits albumin to carry paclitaxel directly into cancer cells through a receptor-utilized carrier system, enhancing paclitaxel distribution at the tumor site [16]. As graphically shown in Figure 3, the binding of albumin with the gp60 receptor stimulates caveolin-1, resulting in the formation of vesicles known as caveolin, which facilitate the transport of the albumin-drug complex into the tumor interstitial space through transcytosis [16]. Consequently, there is an enhanced intratumoral nab-paclitaxel accumulation due to its enhanced binding and transport [9]. Once nab-paclitaxel is in the extravascular space, it can accumulate inside the tumor through the interaction between SPARC (secreted protein acidic and rich in cysteine) and albumin. SPARC is present in the tumor microenvironment of many cancers including gastrointestinal cancers [17,18] (Figure 3). It has been demonstrated that SPARC promotes intratumoral aggregation of albumin-bound nanoparticles [19], and nab-paclitaxel utilizes this characteristic of cancer biology, as it is preferentially retained by tumoral SPARC.
This article provides a review of nab-paclitaxel’s role in improving clinical efficacy and safety during the treatment of gastrointestinal (GI) cancers.

2. Pharmacokinetics of Nab-Paclitaxel

Nab-paclitaxel has been shown to improve the pharmacokinetics and biodistribution of paclitaxel compared to solvent-based paclitaxel (sb-paclitaxel) in solid cancers [20,21]. In these studies, the maximum plasma concentration (Cmax) of paclitaxel was 3.8 to 6.5 folds higher after infusion of nab-paclitaxel compared to sb-paclitaxel [20,21]. In phase I clinical trials, the plasma pharmacokinetics (PK) of paclitaxel or gemcitabine were compared between nab-paclitaxel (125 mg/m2) monotherapy and nab-paclitaxel (125 mg/m2) plus gemcitabine (1000 mg/m2) combination therapy [22,23]. The results showed no significant differences in peak plasma concentration (Cmax), clearance (CL), volume distribution (Vz) or half-life (t1/2) of either paclitaxel or gemcitabine between the treatments [22,23]. In another phase I pharmacokinetics study, where nab-paclitaxel was used in combination with oral fluoropyridine S-1 [24], the PK parameters of the drugs were very similar when nab-paclitaxel was administered either alone or co-administered with S-1 [24]. These results indicate that nab-paclitaxel does not have any significant drug interactions when used in combination with other anticancer therapies. In another phase I PK study, nab-paclitaxel was administered in patients with solid tumors, including GI cancers, through hepatic atrial infusion (HAI) to compare the hepatic extraction of nab-paclitaxel between hepatic arterial and intravenous infusions [24]. Hepatic extraction of nab-paclitaxel represents the amount of nab-paclitaxel directly released to the liver, bypassing systemic vulnerability. In this study, approximately 42% higher hepatic extraction of nab-paclitaxel was observed following HAI compared to intravenous infusion, indicating that HAI can achieve better antitumor activity in patients with liver metastasis [24].

3. Nab-Paclitaxel in Pancreatic Cancer

Pancreatic ductal adenocarcinoma (PDAC) has the highest mortality rate among all major cancers. PDAC has a poor prognosis with a 5-year survival rate of <10% for all stages combined [25]. Gemcitabine (Gem) has remained the standard treatment for advanced PDAC patients since 1997; however, it only demonstrated a modest clinical response with a median survival of approximately 6 months [26,27]. The combination chemotherapy regimen FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan, oxaliplatin) improved the survival of PDAC patients to 11.1 months, compared to 6.8 months with gemcitabine. However, this regimen resulted in increased toxicity [28]. Solvent-based taxanes, docetaxel or paclitaxel, either alone or in combination with gemcitabine, have only shown moderate clinical activity [29,30,31,32,33,34,35]. Awasthi et al. demonstrated the superior activity of nab-paclitaxel compared with gemcitabine or docetaxel and showed improved activity of a nab-paclitaxel plus gemcitabine (nabP/Gem) in PDAC preclinical models [36]. Similarly, the superior therapeutic efficacy of nab-paclitaxel over cremophor solvent-based paclitaxel was observed by others in an orthotopic model of PDAC [37].
Based on the promising clinical activity and improved safety of nab-paclitaxel in breast cancer [38], it was evaluated in PDAC clinical trials. In a phase I/II trial, nabP/Gem showed tolerable adverse effects with substantial antitumor activity, with an overall response rate (ORR) of 48%, median progression-free survival (PFS) of 7.9 months, and median overall survival (OS) of 12.2 months, as first-line therapy in metastatic PDAC patients [39]. In a phase II trial of gemcitabine-refractory advanced PDAC patients, nab-paclitaxel was well tolerated and demonstrated preliminary clinical activity as second-line therapy (ORR 58%, PFS 1.7 months, OS 7.3 months) [40]. A phase III landmark trial (MPACT) comparing the nabP/Gem regimen versus gemcitabine monotherapy in metastatic PDAC patients demonstrated that nabP/Gem was superior to gemcitabine in terms of OS (8.5 vs. 6.7 months), PFS (5.5 vs. 3.7 months), and ORR (23 vs. 7%). These results led to the approval of nabP/Gem as a first-line treatment for metastatic PDAC patients [41]. A phase II study with nabP/Gem vs. nab-paclitaxel plus leucovorin/5-FU showed a PFS rate of 54% vs. 56% after 4 months with a tolerable toxicity profile [42]. A phase II study of untreated metastatic PDAC patients with nabP/Gem plus the HSP27-targeting antisense oligonucleotide apatorsen or placebo exhibited no clinical benefit (PFS 2.7 vs. 3.8 months; OS 5.3 vs. 6.9 months) [43]. In a phase Ib/II study of metastatic PDAC patients, nabP/Gem in combination with pembrolizumab, a PD-1 inhibitor, demonstrated that the regimen was well tolerated and there was a slight improvement in efficacy (PFS 9.1 months; OS 15 months) [44].
Considering the excessive hyaluronan (HA) accumulation in the PDAC tumor microenvironment, pegvorhyaluronidase alfa (PEGPH20), which degrades HA, was evaluated in combination with nabP/Gem. In a phase II trial, this regimen compared to nabP/Gem plus placebo demonstrated improved PFS with the largest improvement in patients with HA-high tumors (PFS 9.2 vs. 5.2 months; OS 11.5 vs. 8.5 mo) [45]. However, in the phase III trial, this regimen increased the ORR (47 vs. 36%) but did not improve OS (11.2 vs. 11.5 months) or PFS (7.1 vs. 7.1 months) [46]. In a single-arm phase II study in advanced PDAC patients, nab-paclitaxel plus S1 followed by S-1 maintenance therapy demonstrated encouraging ORR (53.1%), PFS (6.2 months), OS (13.6 months) and manageable toxicity as first-line therapy [47]. The addition of cisplatin and capecitabine to the nabP/Gem backbone in a phase II trial yielded promising clinical efficacy (6-month disease-free survival rate of 74% vs. 46%) with manageable toxicity [48]. Furthermore, a phase I trial of nabP/Gem in combination with enzalutamide, a novel androgen receptor, exhibited an OS of 9.73 months and PFS of 7.53 months, without any unexpected toxicities [49]. A phase II trial of nabP/Gem in combination with hydroxychloroquine (HCQ), an autophagy inhibitor, in untreated advanced PDAC patients showed no improvement in the primary endpoint of OS at 12 months [50]. However, in a later phase II study of PDAC patients, treatment with preoperative HCQ plus nabP/Gem followed by resection resulted in a greater pathologic tumor response, improved serum biomarker response and evidence of autophagy inhibition and immune activity [51]. In phase II trials, the addition of the Notch signaling inhibitor tarextumab [52], or the insulin-like growth factor-1 receptor and HER3 pathway dual inhibitor istiratumab [53], to nabP/Gem in untreated metastatic PDAC patients did not exhibit any significant clinical improvement. Similarly, the addition of vismodegib, a hedgehog (Hh) signaling inhibitor, to nabP/Gem did not improve clinical efficacy in patients with untreated metastatic PDAC (PFS 5.4 months; OS 9.8 months) [54]. A phase II study of untreated metastatic PDAC patients with nabP/Gem plus the heparin mimetic necuparanib or placebo demonstrated no significant benefit (PFS 5.5 vs. 6.9 months; OS 10.7 vs. 9.9 months) [55]. A phase II study of perioperative nabP/Gem for resectable PDAC showed that it was tolerable, but the primary endpoint of an 85% complete resection rate was not met [56]. As an alternative to FOLFIRINOX, nab-paclitaxel was evaluated in combination with FOLFOX in PDAC patients following surgical resection in phase I [57] and phase II [58] trials. This regimen showed manageable toxicity and significant clinical efficacy with a median disease-free survival of 19.7 months and median OS of 53.5 months [58].
Nivolumab, an immune checkpoint inhibitor (ICI) antibody, in combination with nabP/Gem in advanced PDAC, showed a manageable safety profile without any significant clinical benefit (PFS 5.5 months; OS 9.9 months) in a phase I study [59]. Later, a combination of an agonistic CD40 antibody APX005M (sotigalimab) with nabP/Gem and nivolumab demonstrated good tolerability and clinical activity in a phase Ib study, indicating the potential for this regimen to improve chemotherapy-only clinical activity [60]. Since matrix metalloproteinase 9 (MMP9) expression in the PDAC tumor microenvironment has multiple protumorigenic effects [61], and an anti-MMP9 antibody has antitumor activity in combination with nab-paclitaxel-based chemotherapy in preclinical PDAC models [62], the MMP9 antibody andecaliximab (GS-5745) was evaluated in combination with nabP/Gem in patients with advanced PDAC in a phase I study. This regimen demonstrated a favorable safety profile and clinical activity (PFS 7.8 months; ORR 44.4%) [63]. A phase II study of nabP/Gem versus nabP/Gem followed by FOLFIRINOX induction chemotherapy exhibited similar activity (surgical conversion rate 35.9% vs. 43.9%; OS 18.5 vs. 20.7 months) and safety between the two regimens in locally advanced resectable PDAC patients [64]. A phase III study of nabP/Gem plus ibrutinib, a Bruton’s tyrosine kinase inhibitor, did not improve OS or PFE compared to nabP/Gem in advanced PDAC patients [65]. A phase I/II study of two modified regimens of FOLFIRINOX, replacing either oxaliplatin (Nab-FOLFIRI) or irinotecan (Nab-FOLFOX) with nab-paclitaxel, showed promising clinical activity (PFS 6 vs. 5.6 months; OS 10.2 vs. 10.4 months) in patients with metastatic PDAC [66]. A phase II study of nabP/Gem combination with S1 as neoadjuvant chemotherapy in borderline resectable PDAC with arterial contact demonstrated a 43% response rate, and 96% patients received pancreatectomy. The median PFS and OS were 24.2 and 41 months, respectively [67]. In a phase III trial, algenpantucel-L immunotherapy did not improve survival in patients with borderline resectable or locally advanced unresectable PDAC receiving standard neoadjuvant chemotherapy (FOLFIRINOX or nabP/Gem) and chemoradiation [68]. A phase I/II trial evaluated RX-3117, a small molecule antimetabolite, in combination with nab-paclitaxel and demonstrated tolerability, safety and preliminary efficacy (ORR 23.1%, PFS ~5.6 months) in newly diagnosed metastatic PDAC patients [69]. In a phase II trial, sotigalimab and/or nivolumab were evaluated in combination with nabP/Gem in metastatic PDAC patients. The primary endpoint of 1-year OS was met for nivo/chemo (57.7%, p = 0.006 compared to a historic 1-year OS of 35%) but was not met for sotiga/chemo (48.1%, p = 0.062) or sotiga/nivo/chemo (41.3%, p = 0.223) [70]. A phase II trial of nabP/Gem with or without ICIs durvalumab (PD-L1 antibody) and tremelimumab (CTLA-4 antibody) showed no improvement in survival (PFS 5.5 vs. 5.4 months; OS 9.8 vs. 8.8 months) in metastatic PDAC patients [71]. A phase I trial of dual αV-integrin and neuropilin-1 targeting peptide CEND-1 in combination with nabP/Gem for the treatment of untreated metastatic PDAC patients demonstrated an acceptable safety profile, with no dose-limiting toxicities and encouraging activity (median OS 13.2 months) [72]. A phase II trial of neoadjuvant nabP/Gem for borderline resectable PDAC with arterial involvement showed the safety and effectiveness of this regimen, providing a chance for curative resection and improved survival [73]. A phase Ib study of ulixertinib, a novel ERK inhibitor, with nabP/Gem in untreated metastatic PDAC showed potential efficacy (median PFS 5.46 months; OS 12.23 months), a similar frequency of grade ≥ 3 toxicities, but a high rate of all-grade toxicities [74]. Recently, a phase II trial of mFOLFIRINOX versus nabP/Gem in advanced PDAC reported nabP/Gem as a better regimen than mFOLFIRINOX because of its better RR (42.1% vs. 30.9%), CA19-9 response rate (85% vs. 57.1%) and mild gastrointestinal toxicities. Both regimens displayed higher efficacy in the 1-year OS (82.5% and 77.4%) than in the historic data of gemcitabine monotherapy [75]. Phase II and III clinical trials of nab-paclitaxel in PDAC are summarized in Table 1.
Mechanisms of chemoresistance in PDAC involve the heterogeneous stroma, consisting of fibroblasts, myofibroblasts, immune cells, blood vessels, extracellular matrix (ECM), and soluble proteins. The dense desmoplastic stroma acts as a physical barrier for drug delivery, exacerbated by its hypovascular nature [76]. Factors such as pancreatic stellate cells (PaSC), SPARC, hyaluronan, Hedgehog signaling, cytokines, and growth factors contribute to stromal density and hinder drug delivery [77,78,79,80]. PDAC cells produce angiogenic and anti-angiogenic factors, including angiostatin, endostatin, and pigment epithelium-derived factor, which strongly inhibit angiogenesis [81]. Strategies targeting the fibrotic stroma to reverse hypovascularity and enhance drug delivery have been studied. While some stromal-targeted therapies improve drug delivery and patient survival [78,82], trials focused on the Hedgehog signaling pathway showed no clinical benefits [83]. Additionally, hypoxia, a common feature of PDAC, enhances resistance to cytotoxic drugs by slowing the cell cycle and activating hypoxia-inducible factor-1 (HIF-1) to promote cancer cell survival [81]. In the PDAC tumor microenvironment (TME), exosomes play a critical role in chemoresistance, with cancer-associated fibroblasts and cancer stem cells transferring resistance via exosomes [84,85]. Moreover, PDACs can acquire chemoresistance through the uptake of exosomes. At the molecular level, gemcitabine relies on human nucleoside transporters (NTs), particularly hENT1, for effective transport into tumor cells. Deficiency or inhibition of hENT1 leads to gemcitabine resistance [86]. Enzymatic processes, including phosphorylation, deamination, and dephosphorylation, influence gemcitabine’s efficacy, with enzymes like cytidine deaminase (CDA), deoxycytidylate deaminase (dCTD), and 5′-nucleotidases (5′-NTs) affecting its metabolism. These molecular mechanisms impact gemcitabine’s cytotoxicity and overall effectiveness [80,87]. Understanding these mechanisms is crucial for developing strategies to overcome chemoresistance in PDAC.
The chemoresistance mechanisms of nab-paclitaxel in PDAC are similar to those of paclitaxel, and its use in various types of cancer suggests several possible mechanisms. Several major proteins involved in multidrug resistance, including P-glycoprotein (P-gp), also known as multidrug resistance protein 1 (MDR1), MDR-associated protein 1 (MRP1), and breast cancer resistance protein (BCRP), are involved in resistance to all taxanes [88,89]. The impact of paclitaxel on microtubules can lead to alterations in the β-tubulin family within resistant cancer cells. Notably, overexpression of β-tubulin III in PDAC has been linked to increased tumor growth and metastasis [90,91]. Additionally, the gene HE4 (human epididymis protein 4) has been shown to promote PDAC cell growth and reduce sensitivity to paclitaxel, indicating that HE4 expression may be used to predict the sensitivity of PDAC patients to paclitaxel [92]. Recent findings have revealed that sustained induction of c-MYC is associated with nab-paclitaxel resistance in primary PDAC cells [93]. Moreover, exosomes have been shown to influence multidrug resistance proteins involved in paclitaxel resistance [94].

4. Nab-Paclitaxel in Esophageal Cancer

There are two main subtypes of esophageal cancer (EC): esophageal adenocarcinoma (EAD) and esophageal squamous cell carcinoma (ESCC) [95]. ESCC, which is more common in Asian countries, accounts for about 90% of the EC worldwide, whereas EAC is the leading type of EC in the USA [96]. In our preclinical studies of esophageal cancer, we observed a higher effectiveness for nab-paclitaxel compared to sb-paclitaxel [97]. As first-line therapy, nab-paclitaxel’s efficacy and safety were compared in a retrospective study where nab-paclitaxel with cisplatin and sb-paclitaxel with cisplatin were given in metastatic ESCC patients [98]. In this study, 32 patients received two cycles of nab-paclitaxel (125 mg/m2) plus cisplatin (75 mg/m2) (Nab-TP group) intravenously over a 30-min period with adequate hydration but without any premedication in a 21-day period. Similarly, 43 patients received two cycles of sb-paclitaxel (80 mg/m2) and the same dose of cisplatin (sb-TP group) for the same 21-day period with premedication of corticosteroids and antihistamines. The incidence of grade ≥3 peripheral neuropathy, arthralgia, and myalgia were significantly lower in the nab-TP group compared to the sb-TP group (all p < 0.05), indicating that nab-TP was generally better tolerated. The incidence of hematological toxicities like anemia, leukopenia, neutropenia, febrile neutropenia, and thrombocytopenia did not differ significantly between the two study arms. However, the comparative efficacy and safety profiles of this study were consistent with those in other reports [99,100,101] where nab-paclitaxel was used as the first-line treatment for metastatic ESCC [99,101]. Additionally, nab-TP demonstrated a higher ORR (50% vs. 30%; p = 0.082) and disease control rate (DCR) (81% vs. 65%; p = 0.124) than sb-TP. Although the median OS was similar between these two groups, nab-TP resulted in a longer median PFS (6.1 months (95% confidence interval: 5.3–6.9)) than sb-TP (5.0 months (95% confidence interval: 4.4–5.6)) (p = 0.029).
The therapeutic applicability of nab-paclitaxel, along with its efficacy and safety profiles, have been tested in a variety of clinical studies which are listed in Table 2. Neoadjuvant chemotherapies and chemoradiotherapies have been used as the standard of care for the treatment of advanced EC [102]. The use of nab-paclitaxel in combination with platinum-based therapy, such as cisplatin, has also been explored as neoadjuvant therapy in locally advanced ESSC [103]. In this phase II study using nab-paclitaxel plus cisplatin as neoadjuvant therapy, the ORR was 65.7% and the median OS was 37.8 months, demonstrating the promising role of nab-paclitaxel as an alternative treatment [103]. In this study, no treatment-related death was reported among patients. When comparing treatment-related toxicities, grade ≥ 3 neutropenia was higher (61.9% vs. 11.5%) with a greater risk of postoperative death in a comparable study where sb-paclitaxel was used instead of nab-paclitaxel [104]. The therapeutic efficacy and safety of nab-paclitaxel in combination with ICIs have recently been evaluated as neoadjuvant therapies for advanced EC [105,106,107,108,109]. In these studies, patients received preoperative nab-paclitaxel in combination with ICIs such as camrelizumab [105,106,108,109,110] or tislelizumab [107]. Some treatment-related adverse events (TRAEs) were reported in these studies, but none of them were previously unreported toxicities. Additionally, very few or no patients experienced grade 3 or higher TRAEs. There were no treatment-related perioperative or postoperative deaths reported. A very strong antitumor response was observed with nab-paclitaxel plus camrelizumab, as evidenced by more than 20% to 50% of patients having a complete pathological response. There was also evidence of the complete disappearance of the primary tumors in a significant number of cases, with an enhanced ORR in the nab-paclitaxel plus camrelizumab arms. When comparing with sb-paclitaxel, the complete pathological response was significantly higher in the ICIs plus nab-paclitaxel group compared to the ICIs plus sb-paclitaxel group (36.7% vs. 21.4%, p = 0.018) [109].

5. Nab-Paclitaxel in Gastric Cancer

Gastric adenocarcinoma (GAC) is the fifth most common cancer and the fourth leading cause of cancer-related death worldwide [96]. For unresectable metastatic or recurrent GAC, combination chemotherapy regimens demonstrate a dismal clinical benefit, leading to a median survival of less than a year [121,122,123,124]. The FLOT (5-FU/leucovorin, oxaliplatin and docetaxel) regimen demonstrated better OS (50 months) compared with the ECF/ECX (epirubicin, cisplatin, fluorouracil or capecitabine) group (35 months) as a perioperative therapy for GAC patients with locally advanced, resectable tumors [125]. Thus, the FLOT regimen is now the standard regimen for a perioperative strategy of resectable GAC patients and is widely utilized for metastatic GAC as well. Meta-analyses indicate that GAC patients’ survival can be improved by second-line therapy after failing first-line chemotherapy [126,127]. Second-line therapy for GAC patients usually involves cytotoxic chemotherapy agents such as taxanes and irinotecan or the two molecular targeted agents trastuzumab and ramucirumab [128]. The median OS of GAC patients receiving second-line therapy ranges from 3.6 to 10.9 months [112,129,130]. In GAC preclinical models, Awasthi et al. recently demonstrated that nab-paclitaxel and liposomal irinotecan (nal-IRI) have higher anti-tumor efficacy than their solvent-based formulations [131,132,133,134,135]. Due to the low response rates of current standard therapies, and the development of chemoresistance and toxicity [136], several clinical studies evaluated nab-paclitaxel in GAC (Table 2).
A phase II study of nab-paclitaxel (given every 3 weeks) showed promising activity in previously treated unresectable or recurrent GAC (ORR 28 = 7.8%, PFS 2.9 months, OS 9.2 months), with well-tolerated toxicities [111]. A phase I study of nab-paclitaxel in combination with S-1 in unresectable or recurrent GAC patients demonstrated preliminary clinical activity with good tolerability [24]. A phase III trial evaluated nab-paclitaxel versus sb-paclitaxel in previously treated advanced GAC patients. This trial showed that weekly nab-paclitaxel was not inferior to weekly sb-paclitaxel in terms of OS and recommended nab-paclitaxel as a potential second-line GAC therapy based on the other advantages associated with this regimen [112]. A phase II study of nab-paclitaxel plus ramucirumab in previously treated advanced GAC patients demonstrated promising activity with manageable toxicities. In this trial, the ORR was 54.8% and PFS was 7.6 months [112]. A phase II study of tri-weekly low-dose nab-paclitaxel (180 mg/m2) showed clinical efficacy (ORR 5.9%, median PFS 2.4 months and OS 9.2 months) in advanced GAC patients with good tolerability [113]. A phase III trial showed that peritoneal metastasis might be a predictive factor for nab-paclitaxel second-line treatment of advanced GAC patients [114]. A triple chemotherapy combination of nab-paclitaxel, oxaliplatin and 5-FU as a perioperative regimen in resectable GAC patients demonstrated promising activity with 95.5% of patients having R0 tumor resection, but there was a significant rate of surgical complications [137]. A phase II trial of nab-paclitaxel as a second-line therapy for fluoropyrimidine-refractory advanced GAC patients demonstrated a manageable safety profile and significant clinical activity with a median PFS of 3.5 months, OS of 9 months, ORR of 16% and DCR of 72% [115]. In Japanese patients, a reduced dose of nab-paclitaxel (tri-weekly 220 mg/m2) was not recommended as a second-line therapy in advanced or recurrent GAC based on its dismal response rate [116].

6. Nab-Paclitaxel in Cholangiocarcinoma

Cholangiocarcinoma (CCA) is a highly aggressive biliary tract cancer (BTC). Depending on their anatomical site of origin, CCAs are divided into three subtypes: intrahepatic (iCCA, 10–20%), perihilar (pCCA, 50–60%), and distal (dCCA, 20–30%) [138]. CCA has a very poor prognosis with a 5-year survival rate of 5–15% [139]. Complete surgical resection is the only chance for long-term survival, but unfortunately, most CCAs are diagnosed with metastatic or locally advanced unresectable disease [140]. For unresectable and recurrent CCA, combination chemotherapy with gemcitabine and cisplatin (GemCis) remains the standard treatment despite moderate clinical activity, as demonstrated by the median survival of only 14 months [141]. Nab-paclitaxel is currently under clinical investigation for CCA based on its superior antitumor efficacy and safety profile in other solid tumors (Table 2).
Nab-paclitaxel plus gemcitabine was evaluated as a first-line treatment of advanced and metastatic CCA in a phase II trial. This regimen was well tolerated but did not meet its primary efficacy endpoint [117]. Considering historical data on PFS of 8.0 months and an OS of 11.7 months with the GemCis regimen [142], a phase II trial of gemcitabine, cisplatin and nab-paclitaxel combination demonstrated significant clinical activity by prolonging PFS (11.8 months) and OS (19.2 months) [118]. A phase III trial of this regimen in treating patients with newly diagnosed advanced CCA is currently ongoing (NCT03768414). A single-arm phase I study of nab-paclitaxel plus capecitabine as a second-line therapy in GemCis refractory advanced CCA patients demonstrated adequate safety and promising early efficacy with median PFS and OS of 5.7 and 12.1 months, respectively [143].

7. Nab-Paclitaxel in Colorectal Cancer

Colorectal cancer (CRC) is the third biggest cause of cancer-associated death and the fourth most frequently diagnosed cancer in the world [144]. Even though taxanes are widely used for the treatment of upper GI tract (esophageal and gastric) cancers, their use in the treatment of lower GI tract (colorectal) cancers is very limited due to poor response and/or development of resistance, although the mechanism of this resistance is not clearly known [145]. Resistance to paclitaxel in CRC can be related to P-gp, as it is overexpressed in CRC [146,147], and a higher dose of paclitaxel has been associated with overcoming the paclitaxel resistance [148]. Thus, there is a therapeutic potential of using nab-paclitaxel to overcome paclitaxel resistance in CRC, as an enhanced accumulation of nab-paclitaxel can be achieved inside CRC xenografts compared to sb-paclitaxel [9]. In addition, the enhanced in vivo antitumor effect of nab-paclitaxel, overcoming sb-paclitaxel resistance, is attributed to its antiangiogenic mechanism [149]. However, a multicenter phase II clinical trial of nab-paclitaxel monotherapy in previously treated metastatic CRC (mCRC) patients did not show an encouraging anticancer effect [119]. Interestingly, in another phase II clinical trial, nab-paclitaxel monotherapy exhibited improvement in median PFS in small bowel adenocarcinoma compared to CRC having a high frequency of genome methylation at CpG islands (CIMP-high CRC) (3.2 months vs. 2.1 months, p = 0.03) [120]. Taxane resistance has been linked to CIMP-high CRC patients [150], and nab-paclitaxel monotherapy did not improve clinical outcomes in these patients, suggesting that CIMP-high should not be used as a predictive biomarker for nab-paclitaxel monotherapy. On the other hand, nab-paclitaxel at a dose of 200 mg/m2 for 21 days proved to be safe in intestinal cancer patients. Adverse effects were reported in less than 10% of the patients, with no report of grade 5 adverse effects. Thus, nab-paclitaxel monotherapy may not be an effective therapeutic option for treating mCRC, and nab-paclitaxel in combination with another anticancer drug may be more effective. Results from ongoing or recently completed clinical trials (NCT03563157, NCT01730586, NCT02574663, NCT02857270) where nab-paclitaxel has been used in combination with other chemotherapies or targeted therapies for the treatment of CRC may provide answers regarding the role of nab-paclitaxel in CRC combination therapies. The comparative role of nab-paclitaxel as mono and combination therapies in CRC has been summarized in Table 2.

8. Future Prospective and Conclusions

Nab-paclitaxel has shown superior activity to paclitaxel in many types of cancers, including GI cancers. However, clinical response to nab-paclitaxel can be hampered by resistance development, and the associated mechanism is not well studied. SPARC is a secreted protein that is overexpressed in multiple cancers, including GI cancers, and its overexpression may be associated with a positive response to nab-paclitaxel [151]. As SPARC is an albumin-binding protein, it is thought to facilitate the intratumoral accumulation of nab-paclitaxel [9]. However, the role of SPARC as a predictive biomarker in GI cancers is not very clear [152]. Therefore, further studies are needed to determine the potential role of novel predictive biomarkers in assessing the nab-paclitaxel response in GI cancers.
The nanoparticle albumin-bound treatment strategy is an encouraging method for delivering other water-insoluble anticancer drugs. Drugs like docetaxel and the mTOR (mammalian target of rapamycin) inhibitor rapamycin have been formulated as nanoparticles in solvent-free drug formulations. A phase I clinical trial (NCT04931823) is currently recruiting patients to study the safety of nab-docetaxel (CPO-100) in advanced solid tumors. Similarly, nab-sirolimus (nab-rapamycin) is currently being investigated in a phase I clinical trial (NCT05661461) involving various solid tumors. These novel nanoparticle albumin-bound drugs will hopefully display higher tumor penetration and anti-tumor activity in various cancers, including GI cancers.
In conclusion, nab-paclitaxel has demonstrated improved clinical activity and safety compared to sb-paclitaxel in most GI cancers. While nab-paclitaxel is currently approved for metastatic PDAC among all GI cancers, its efficacy and safety in other GI cancers are being evaluated in clinical trials. Based on encouraging results from these clinical trials, the utilization of nab-paclitaxel in different GI cancers is expected to increase in the future. Thus, nab-paclitaxel has the potential to become part of clinical regimens to improve the therapy of most GI cancer patients.

Author Contributions

Conceptualization, M.S.H., N.A. and U.v.H.; methodology, M.S.H., N.A., S.P. and U.v.H.; software, M.S.H., N.A., S.P. and U.v.H.; validation, M.S.H., N.A., S.P. and U.v.H.; formal analysis, M.S.H.; investigation, M.S.H.; resources M.S.H., N.A. and U.v.H.; data curation, M.S.H. and N.A.; writing—original draft preparation, M.S.H. and N.A.; writing—review and editing, N.A. and U.v.H.; visualization, M.S.H.; supervision, M.S.H.; project administration, M.S.H.; funding acquisition, M.S.H. and U.v.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Indiana University School of Medicine internal funding and the Goshen Center for Cancer Care funding.

Institutional Review Board Statement

The study was approved by the Indiana University School of Medicine Institutional Biosafety Committee (IBC Protocol # SM-1283).

Informed Consent Statement

Not Applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no cometing interest.

References

  1. Menzin, A.W.; King, S.A.; Aikins, J.K.; Mikuta, J.J.; Rubin, S.C. Taxol (paclitaxel) was approved by FDA for the treatment of patients with recurrent ovarian cancer. Gynecol. Oncol. 1994, 54, 103. [Google Scholar]
  2. Tannock, I.F.; de Wit, R.; Berry, W.R.; Horti, J.; Pluzanska, A.; Chi, K.N.; Oudard, S.; Théodore, C.; James, N.D.; Turesson, I.; et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. Med. 2004, 351, 1502–1512. [Google Scholar] [CrossRef] [Green Version]
  3. Sparreboom, A.; van Zuylen, L.; Brouwer, E.; Loos, W.J.; de Bruijn, P.; Gelderblom, H.; Pillay, M.; Nooter, K.; Stoter, G.; Verweij, J. Cremophor EL-mediated alteration of paclitaxel distribution in human blood: Clinical pharmacokinetic implications. Cancer Res. 1999, 59, 1454–1457. [Google Scholar] [PubMed]
  4. ten Tije, A.J.; Verweij, J.; Loos, W.J.; Sparreboom, A. Pharmacological effects of formulation vehicles: Implications for cancer chemotherapy. Clin. Pharmacokinet. 2003, 42, 665–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gallo, J.M.; Li, S.; Guo, P.; Reed, K.; Ma, J. The effect of P-glycoprotein on paclitaxel brain and brain tumor distribution in mice. Cancer Res. 2003, 63, 5114–5117. [Google Scholar] [PubMed]
  6. Hendrikx, J.J.; Lagas, J.S.; Rosing, H.; Schellens, J.H.; Beijnen, J.H.; Schinkel, A.H. P-glycoprotein and cytochrome P450 3A act together in restricting the oral bioavailability of paclitaxel. Int. J. Cancer 2013, 132, 2439–2447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Stinchcombe, T.E. Nanoparticle albumin-bound paclitaxel: A novel Cremphor-EL-free formulation of paclitaxel. Nanomedicine 2007, 2, 415–423. [Google Scholar] [CrossRef]
  8. Gradishar, W.J. Albumin-bound paclitaxel: A next-generation taxane. Expert. Opin. Pharmacother. 2006, 7, 1041–1053. [Google Scholar] [CrossRef]
  9. Desai, N.; Trieu, V.; Yao, Z.; Louie, L.; Ci, S.; Yang, A.; Tao, C.; De, T.; Beals, B.; Dykes, D.; et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin. Cancer Res. 2006, 12, 1317–1324. [Google Scholar] [CrossRef] [Green Version]
  10. Ibrahim, N.K.; Desai, N.; Legha, S.; Soon-Shiong, P.; Theriault, R.L.; Rivera, E.; Esmaeli, B.; Ring, S.E.; Bedikian, A.; Hortobagyi, G.N.; et al. Phase I and pharmacokinetic study of ABI-007, a Cremophor-free, protein-stabilized, nanoparticle formulation of paclitaxel. Clin. Cancer Res. 2002, 8, 1038–1044. [Google Scholar]
  11. Borgå, O.; Henriksson, R.; Bjermo, H.; Lilienberg, E.; Heldring, N.; Loman, N. Maximum Tolerated Dose and Pharmacokinetics of Paclitaxel Micellar in Patients with Recurrent Malignant Solid Tumours: A Dose-Escalation Study. Adv. Ther. 2019, 36, 1150–1163. [Google Scholar] [CrossRef] [Green Version]
  12. FDA Approves Abraxane for Metastatic Breast Cancer. Oncology NEWS International; MJH Life Sciences: Cranbury, NJ, USA, 2005. [Google Scholar]
  13. Levin, J. FDA Approves ABRAXANE® for the First-Line Treatment of Advanced Non-Small Cell Lung Cancer; Fierce Pharma: Washington, DC, USA, 2012. [Google Scholar]
  14. Kundranda, M.N.; Niu, J. Albumin-bound paclitaxel in solid tumors: Clinical development and future directions. Drug Des. Dev. Ther. 2015, 9, 3767–3777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hughes, G.A. Nanostructure-mediated drug delivery. Nanomedicine 2005, 1, 22–30. [Google Scholar] [CrossRef]
  16. Foote, M. Using nanotechnology to improve the characteristics of antineoplastic drugs: Improved characteristics of nab-paclitaxel compared with solvent-based paclitaxel. Biotechnol. Annu. Rev. 2007, 13, 345–357. [Google Scholar] [CrossRef]
  17. Desai, N.P.; Trieu, V.; Hwang, L.Y.; Wu, R.; Soon-Shiong, P.; Gradishar, W.J. Improved effectiveness of nanoparticle albumin-bound (nab) paclitaxel versus polysorbate-based docetaxel in multiple xenografts as a function of HER2 and SPARC status. Anticancer Drugs 2008, 19, 899–909. [Google Scholar] [CrossRef] [PubMed]
  18. Nagaraju, G.P.; El-Rayes, B.F. SPARC and DNA methylation: Possible diagnostic and therapeutic implications in gastrointestinal cancers. Cancer Lett. 2013, 328, 10–17. [Google Scholar] [CrossRef] [PubMed]
  19. Trieu, V.; Hwang, J.; Desai, N. Nanoparticle Albumin-bound (nab) technology may enhance antitumor activity via targeting of SPARC protein. Abstract 53; New Targets and Delivery System for Cancer Diagnosis and Treatment (SKCC): San Diego, CA, USA.
  20. Gardner, E.R.; Dahut, W.L.; Scripture, C.D.; Jones, J.; Aragon-Ching, J.B.; Desai, N.; Hawkins, M.J.; Sparreboom, A.; Figg, W.D. Randomized crossover pharmacokinetic study of solvent-based paclitaxel and nab-paclitaxel. Clin. Cancer Res. 2008, 14, 4200–4205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Sparreboom, A.; Scripture, C.D.; Trieu, V.; Williams, P.J.; De, T.; Yang, A.; Beals, B.; Figg, W.D.; Hawkins, M.; Desai, N. Comparative preclinical and clinical pharmacokinetics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cremophor (Taxol). Clin. Cancer Res. 2005, 11, 4136–4143. [Google Scholar] [CrossRef] [Green Version]
  22. Ueno, H.; Ikeda, M.; Ueno, M.; Mizuno, N.; Ioka, T.; Omuro, Y.; Nakajima, T.E.; Furuse, J. Phase I/II study of nab-paclitaxel plus gemcitabine for chemotherapy-naive Japanese patients with metastatic pancreatic cancer. Cancer Chemother. Pharmacol. 2016, 77, 595–603. [Google Scholar] [CrossRef]
  23. Cohen, S.J.; O’Neil, B.H.; Berlin, J.; Ames, P.; McKinley, M.; Horan, J.; Catalano, P.M.; Davies, A.; Weekes, C.D.; Leichman, L. A phase 1b study of erlotinib in combination with gemcitabine and nab-paclitaxel in patients with previously untreated advanced pancreatic cancer: An Academic Oncology GI Cancer Consortium study. Cancer Chemother. Pharmacol. 2016, 77, 693–701. [Google Scholar] [CrossRef]
  24. Nakayama, N.; Ishido, K.; Chin, K.; Nishimura, K.; Azuma, M.; Matsusaka, S.; Inokuchi, Y.; Tanabe, S.; Kumekawa, Y.; Koizumi, W. A phase I study of S-1 in combination with nab-paclitaxel in patients with unresectable or recurrent gastric cancer. Gastric Cancer 2017, 20, 350–357. [Google Scholar] [CrossRef] [PubMed]
  25. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  26. Burris, H.A., 3rd; Moore, M.J.; Andersen, J.; Green, M.R.; Rothenberg, M.L.; Modiano, M.R.; Cripps, M.C.; Portenoy, R.K.; Storniolo, A.M.; Tarassoff, P.; et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J. Clin. Oncol. 1997, 15, 2403–2413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Reni, M.; Cordio, S.; Milandri, C.; Passoni, P.; Bonetto, E.; Oliani, C.; Luppi, G.; Nicoletti, R.; Galli, L.; Bordonaro, R.; et al. Gemcitabine versus cisplatin, epirubicin, fluorouracil, and gemcitabine in advanced pancreatic cancer: A randomised controlled multicentre phase III trial. Lancet Oncol. 2005, 6, 369–376. [Google Scholar] [CrossRef]
  28. Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.L.; Gourgou-Bourgade, S.; de la Fouchardière, C.; et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [Green Version]
  29. Okada, S.; Sakata, Y.; Matsuno, S.; Kurihara, M.; Sasaki, Y.; Ohashi, Y.; Taguchi, T. Phase II study of docetaxel in patients with metastatic pancreatic cancer: A Japanese cooperative study. Cooperative Group of Docetaxel for Pancreatic Cancer in Japan. Br. J. Cancer 1999, 80, 438–443. [Google Scholar] [CrossRef] [Green Version]
  30. Rougier, P.; Adenis, A.; Ducreux, M.; de Forni, M.; Bonneterre, J.; Dembak, M.; Clouet, P.; Lebecq, A.; Baille, P.; Lefresne-Soulas, F.; et al. A phase II study: Docetaxel as first-line chemotherapy for advanced pancreatic adenocarcinoma. Eur. J. Cancer 2000, 36, 1016–1025. [Google Scholar] [CrossRef]
  31. Cereda, S.; Reni, M. Weekly docetaxel as salvage therapy in patients with gemcitabine-refractory metastatic pancreatic cancer. J. Chemother. 2008, 20, 509–512. [Google Scholar] [CrossRef]
  32. Saif, M.W.; Syrigos, K.; Penney, R.; Kaley, K. Docetaxel second-line therapy in patients with advanced pancreatic cancer: A retrospective study. Anticancer Res. 2010, 30, 2905–2909. [Google Scholar]
  33. Schneider, B.P.; Ganjoo, K.N.; Seitz, D.E.; Picus, J.; Fata, F.; Stoner, C.; Calley, C.; Loehrer, P.J. Phase II study of gemcitabine plus docetaxel in advanced pancreatic cancer: A Hoosier Oncology Group study. Oncology 2003, 65, 218–223. [Google Scholar] [CrossRef]
  34. Lutz, M.P.; Van Cutsem, E.; Wagener, T.; Van Laethem, J.L.; Vanhoefer, U.; Wils, J.A.; Gamelin, E.; Koehne, C.H.; Arnaud, J.P.; Mitry, E.; et al. Docetaxel plus gemcitabine or docetaxel plus cisplatin in advanced pancreatic carcinoma: Randomized phase II study 40984 of the European Organisation for Research and Treatment of Cancer Gastrointestinal Group. J. Clin. Oncol. 2005, 23, 9250–9256. [Google Scholar] [CrossRef] [PubMed]
  35. Ryan, D.P.; Kulke, M.H.; Fuchs, C.S.; Grossbard, M.L.; Grossman, S.R.; Morgan, J.A.; Earle, C.C.; Shivdasani, R.; Kim, H.; Mayer, R.J.; et al. A Phase II study of gemcitabine and docetaxel in patients with metastatic pancreatic carcinoma. Cancer 2002, 94, 97–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Awasthi, N.; Zhang, C.; Schwarz, A.M.; Hinz, S.; Wang, C.; Williams, N.S.; Schwarz, M.A.; Schwarz, R.E. Comparative benefits of Nab-paclitaxel over gemcitabine or polysorbate-based docetaxel in experimental pancreatic cancer. Carcinogenesis 2013, 34, 2361–2369. [Google Scholar] [CrossRef] [Green Version]
  37. Rajeshkumar, N.V.; Yabuuchi, S.; Pai, S.G.; Tong, Z.; Hou, S.; Bateman, S.; Pierce, D.W.; Heise, C.; Von Hoff, D.D.; Maitra, A.; et al. Superior therapeutic efficacy of nab-paclitaxel over cremophor-based paclitaxel in locally advanced and metastatic models of human pancreatic cancer. Br. J. Cancer 2016, 115, 442–453. [Google Scholar] [CrossRef] [Green Version]
  38. Mahtani, R.L.; Parisi, M.; Glück, S.; Ni, Q.; Park, S.; Pelletier, C.; Faria, C.; Braiteh, F. Comparative effectiveness of early-line nab-paclitaxel vs. paclitaxel in patients with metastatic breast cancer: A US community-based real-world analysis. Cancer. Manag. Res. 2018, 10, 249–256. [Google Scholar] [CrossRef] [Green Version]
  39. Von Hoff, D.D.; Ramanathan, R.K.; Borad, M.J.; Laheru, D.A.; Smith, L.S.; Wood, T.E.; Korn, R.L.; Desai, N.; Trieu, V.; Iglesias, J.L.; et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: A phase I/II trial. J. Clin. Oncol. 2011, 29, 4548–4554. [Google Scholar] [CrossRef] [Green Version]
  40. Hosein, P.J.; de Lima Lopes, G., Jr.; Pastorini, V.H.; Gomez, C.; Macintyre, J.; Zayas, G.; Reis, I.; Montero, A.J.; Merchan, J.R.; Rocha Lima, C.M. A phase II trial of nab-Paclitaxel as second-line therapy in patients with advanced pancreatic cancer. Am. J. Clin. Oncol. 2013, 36, 151–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [Green Version]
  42. Bachet, J.B.; Hammel, P.; Desramé, J.; Meurisse, A.; Chibaudel, B.; André, T.; Debourdeau, P.; Dauba, J.; Lecomte, T.; Seitz, J.F.; et al. Nab-paclitaxel plus either gemcitabine or simplified leucovorin and fluorouracil as first-line therapy for metastatic pancreatic adenocarcinoma (AFUGEM GERCOR): A non-comparative, multicentre, open-label, randomised phase 2 trial. Lancet Gastroenterol. Hepatol. 2017, 2, 337–346. [Google Scholar] [CrossRef]
  43. Ko, A.H.; Murphy, P.B.; Peyton, J.D.; Shipley, D.L.; Al-Hazzouri, A.; Rodriguez, F.A.; Womack, M.S.t.; Xiong, H.Q.; Waterhouse, D.M.; Tempero, M.A.; et al. A Randomized, Double-Blinded, Phase II Trial of Gemcitabine and Nab-Paclitaxel Plus Apatorsen or Placebo in Patients with Metastatic Pancreatic Cancer: The RAINIER Trial. Oncologist 2017, 22, 1427-e1129. [Google Scholar] [CrossRef] [Green Version]
  44. Weiss, G.J.; Blaydorn, L.; Beck, J.; Bornemann-Kolatzki, K.; Urnovitz, H.; Schütz, E.; Khemka, V. Phase Ib/II study of gemcitabine, nab-paclitaxel, and pembrolizumab in metastatic pancreatic adenocarcinoma. Investig. New Drugs 2018, 36, 96–102. [Google Scholar] [CrossRef] [PubMed]
  45. Hingorani, S.R.; Zheng, L.; Bullock, A.J.; Seery, T.E.; Harris, W.P.; Sigal, D.S.; Braiteh, F.; Ritch, P.S.; Zalupski, M.M.; Bahary, N.; et al. HALO 202: Randomized Phase II Study of PEGPH20 Plus Nab-Paclitaxel/Gemcitabine Versus Nab-Paclitaxel/Gemcitabine in Patients With Untreated, Metastatic Pancreatic Ductal Adenocarcinoma. J. Clin. Oncol. 2018, 36, 359–366. [Google Scholar] [CrossRef] [PubMed]
  46. Van Cutsem, E.; Tempero, M.A.; Sigal, D.; Oh, D.Y.; Fazio, N.; Macarulla, T.; Hitre, E.; Hammel, P.; Hendifar, A.E.; Bates, S.E.; et al. Randomized Phase III Trial of Pegvorhyaluronidase Alfa With Nab-Paclitaxel Plus Gemcitabine for Patients With Hyaluronan-High Metastatic Pancreatic Adenocarcinoma. J. Clin. Oncol. 2020, 38, 3185–3194. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, W.; Du, C.; Sun, Y.; Yang, L.; Cui, C.; Jiang, Z.; Wang, C.; Wang, J.; Zhou, A. Nab-paclitaxel plus S-1 as first-line followed by S-1 maintenance for advanced pancreatic adenocarcinoma: A single-arm phase II trial. Cancer Chemother. Pharmacol. 2018, 82, 655–660. [Google Scholar] [CrossRef] [Green Version]
  48. Reni, M.; Zanon, S.; Peretti, U.; Chiaravalli, M.; Barone, D.; Pircher, C.; Balzano, G.; Macchini, M.; Romi, S.; Gritti, E.; et al. Nab-paclitaxel plus gemcitabine with or without capecitabine and cisplatin in metastatic pancreatic adenocarcinoma (PACT-19): A randomised phase 2 trial. Lancet Gastroenterol. Hepatol. 2018, 3, 691–697. [Google Scholar] [CrossRef]
  49. Mahipal, A.; Tella, S.H.; Kommalapati, A.; Goyal, G.; Soares, H.; Neuger, A.; Copolla, D.; Kim, J.; Kim, R. Phase 1 trial of enzalutamide in combination with gemcitabine and nab-paclitaxel for the treatment of advanced pancreatic cancer. Investig. New Drugs 2019, 37, 473–481. [Google Scholar] [CrossRef]
  50. Karasic, T.B.; O’Hara, M.H.; Loaiza-Bonilla, A.; Reiss, K.A.; Teitelbaum, U.R.; Borazanci, E.; De Jesus-Acosta, A.; Redlinger, C.; Burrell, J.A.; Laheru, D.A.; et al. Effect of Gemcitabine and nab-Paclitaxel With or Without Hydroxychloroquine on Patients With Advanced Pancreatic Cancer: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019, 5, 993–998. [Google Scholar] [CrossRef]
  51. Zeh, H.J.; Bahary, N.; Boone, B.A.; Singhi, A.D.; Miller-Ocuin, J.L.; Normolle, D.P.; Zureikat, A.H.; Hogg, M.E.; Bartlett, D.L.; Lee, K.K.; et al. A Randomized Phase II Preoperative Study of Autophagy Inhibition with High-Dose Hydroxychloroquine and Gemcitabine/Nab-Paclitaxel in Pancreatic Cancer Patients. Clin. Cancer Res. 2020, 26, 3126–3134. [Google Scholar] [CrossRef] [Green Version]
  52. Hu, Z.I.; Bendell, J.C.; Bullock, A.; LoConte, N.K.; Hatoum, H.; Ritch, P.; Hool, H.; Leach, J.W.; Sanchez, J.; Sohal, D.P.S.; et al. A randomized phase II trial of nab-paclitaxel and gemcitabine with tarextumab or placebo in patients with untreated metastatic pancreatic cancer. Cancer Med. 2019, 8, 5148–5157. [Google Scholar] [CrossRef] [Green Version]
  53. Kundranda, M.; Gracian, A.C.; Zafar, S.F.; Meiri, E.; Bendell, J.; Algül, H.; Rivera, F.; Ahn, E.R.; Watkins, D.; Pelzer, U.; et al. Randomized, double-blind, placebo-controlled phase II study of istiratumab (MM-141) plus nab-paclitaxel and gemcitabine versus nab-paclitaxel and gemcitabine in front-line metastatic pancreatic cancer (CARRIE). Ann. Oncol. 2020, 31, 79–87. [Google Scholar] [CrossRef]
  54. De Jesus-Acosta, A.; Sugar, E.A.; O’Dwyer, P.J.; Ramanathan, R.K.; Von Hoff, D.D.; Rasheed, Z.; Zheng, L.; Begum, A.; Anders, R.; Maitra, A.; et al. Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma. Br. J. Cancer 2020, 122, 498–505. [Google Scholar] [CrossRef] [PubMed]
  55. O’Reilly, E.M.; Barone, D.; Mahalingam, D.; Bekaii-Saab, T.; Shao, S.H.; Wolf, J.; Rosano, M.; Krause, S.; Richards, D.A.; Yu, K.H.; et al. Randomised phase II trial of gemcitabine and nab-paclitaxel with necuparanib or placebo in untreated metastatic pancreas ductal adenocarcinoma. Eur. J. Cancer 2020, 132, 112–121. [Google Scholar] [CrossRef] [PubMed]
  56. Barbour, A.P.; Samra, J.S.; Haghighi, K.S.; Donoghoe, M.W.; Burge, M.; Harris, M.T.; Chua, Y.J.; Mitchell, J.; O’Rourke, N.; Chan, H.; et al. The AGITG GAP Study: A Phase II Study of Perioperative Gemcitabine and Nab-Paclitaxel for Resectable Pancreas Cancer. Ann. Surg. Oncol. 2020, 27, 2506–2515. [Google Scholar] [CrossRef] [PubMed]
  57. Safran, H.; Charpentier, K.P.; Perez, K.; Mantripragada, K.; Miner, T.; DiPetrillo, T.; Kuritzky, B.; Apor, E.; Bishop, K.; Luppe, D.; et al. FOLFOX+Nab-Paclitaxel (FOLFOX-A) for Advanced Pancreatic Cancer: A Brown University Oncology Research Group Phase I Study. Am. J. Clin. Oncol. 2016, 39, 619–622. [Google Scholar] [CrossRef]
  58. Raufi, A.G.; Breakstone, R.; Leonard, K.; Charpentier, K.; Beard, R.; Renaud, J.; Cavanaugh, L.; Sturtevant, A.; MacKinnon, K.; Almhanna, K.; et al. Adjuvant FOLFOX+Nab-Paclitaxel (FOLFOX-A) for Pancreatic Cancer: A Brown University Oncology Research Group Phase II Study (BrUOG295). Am. J. Clin. Oncol. 2020, 43, 857–860. [Google Scholar] [CrossRef]
  59. Wainberg, Z.A.; Hochster, H.S.; Kim, E.J.; George, B.; Kaylan, A.; Chiorean, E.G.; Waterhouse, D.M.; Guiterrez, M.; Parikh, A.; Jain, R.; et al. Open-label, Phase I Study of Nivolumab Combined with nab-Paclitaxel Plus Gemcitabine in Advanced Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 4814–4822. [Google Scholar] [CrossRef]
  60. O’Hara, M.H.; O’Reilly, E.M.; Varadhachary, G.; Wolff, R.A.; Wainberg, Z.A.; Ko, A.H.; Fisher, G.; Rahma, O.; Lyman, J.P.; Cabanski, C.R.; et al. CD40 agonistic monoclonal antibody APX005M (sotigalimab) and chemotherapy, with or without nivolumab, for the treatment of metastatic pancreatic adenocarcinoma: An open-label, multicentre, phase 1b study. Lancet Oncol. 2021, 22, 118–131. [Google Scholar] [CrossRef]
  61. Bloomston, M.; Zervos, E.E.; Rosemurgy, A.S., 2nd. Matrix metalloproteinases and their role in pancreatic cancer: A review of preclinical studies and clinical trials. Ann. Surg Oncol. 2002, 9, 668–674. [Google Scholar] [CrossRef]
  62. Awasthi, N.; Mikels-Vigdal, A.J.; Stefanutti, E.; Schwarz, M.A.; Monahan, S.; Smith, V.; Schwarz, R.E. Therapeutic efficacy of anti-MMP9 antibody in combination with nab-paclitaxel-based chemotherapy in pre-clinical models of pancreatic cancer. J. Cell Mol. Med. 2019, 23, 3878–3887. [Google Scholar] [CrossRef] [Green Version]
  63. Bendell, J.; Sharma, S.; Patel, M.R.; Windsor, K.S.; Wainberg, Z.A.; Gordon, M.; Chaves, J.; Berlin, J.; Brachmann, C.B.; Zavodovskaya, M.; et al. Safety and Efficacy of Andecaliximab (GS-5745) Plus Gemcitabine and Nab-Paclitaxel in Patients with Advanced Pancreatic Adenocarcinoma: Results from a Phase I Study. Oncologist 2020, 25, 954–962. [Google Scholar] [CrossRef]
  64. Kunzmann, V.; Siveke, J.T.; Algül, H.; Goekkurt, E.; Siegler, G.; Martens, U.; Waldschmidt, D.; Pelzer, U.; Fuchs, M.; Kullmann, F.; et al. Nab-paclitaxel plus gemcitabine versus nab-paclitaxel plus gemcitabine followed by FOLFIRINOX induction chemotherapy in locally advanced pancreatic cancer (NEOLAP-AIO-PAK-0113): A multicentre, randomised, phase 2 trial. Lancet Gastroenterol. Hepatol. 2021, 6, 128–138. [Google Scholar] [CrossRef] [PubMed]
  65. Tempero, M.; Oh, D.Y.; Tabernero, J.; Reni, M.; Van Cutsem, E.; Hendifar, A.; Waldschmidt, D.T.; Starling, N.; Bachet, J.B.; Chang, H.M.; et al. Ibrutinib in combination with nab-paclitaxel and gemcitabine for first-line treatment of patients with metastatic pancreatic adenocarcinoma: Phase III RESOLVE study. Ann. Oncol. 2021, 32, 600–608. [Google Scholar] [CrossRef] [PubMed]
  66. Giommoni, E.; Maiello, E.; Vaccaro, V.; Rondini, E.; Vivaldi, C.; Tortora, G.; Toppo, L.; Giordano, G.; Latiano, T.P.; Lamperini, C.; et al. Activity and Safety of NAB-FOLFIRI and NAB-FOLFOX as First-Line Treatment for metastatic Pancreatic Cancer (NabucCO Study). Curr. Oncol. 2021, 28, 1761–1772. [Google Scholar] [CrossRef] [PubMed]
  67. Kondo, N.; Uemura, K.; Sudo, T.; Hashimoto, Y.; Sumiyoshi, T.; Okada, K.; Seo, S.; Otsuka, H.; Murakami, Y.; Takahashi, S. A phase II study of gemcitabine/nab-paclitaxel/S-1 combination neoadjuvant chemotherapy for patients with borderline resectable pancreatic cancer with arterial contact. Eur. J. Cancer 2021, 159, 215–223. [Google Scholar] [CrossRef] [PubMed]
  68. Hewitt, D.B.; Nissen, N.; Hatoum, H.; Musher, B.; Seng, J.; Coveler, A.L.; Al-Rajabi, R.; Yeo, C.J.; Leiby, B.; Banks, J.; et al. A Phase 3 Randomized Clinical Trial of Chemotherapy With or Without Algenpantucel-L (HyperAcute-Pancreas) Immunotherapy in Subjects With Borderline Resectable or Locally Advanced Unresectable Pancreatic Cancer. Ann. Surg. 2022, 275, 45–53. [Google Scholar] [CrossRef]
  69. Babiker, H.; Schlegel, P.J.; Hicks, L.G.; Bullock, A.J.; Burhani, N.; Mahadevan, D.; Elquza, E.; Borad, M.J.; Benaim, E.; Peterson, C.; et al. A multicenter phase 1/2 study investigating the safety, pharmacokinetics, pharmacodynamics and efficacy of a small molecule antimetabolite, RX-3117, plus nab-paclitaxel in pancreatic adenocarcinoma. Investig. New Drugs 2022, 40, 81–90. [Google Scholar] [CrossRef]
  70. Padrón, L.J.; Maurer, D.M.; O’Hara, M.H.; O’Reilly, E.M.; Wolff, R.A.; Wainberg, Z.A.; Ko, A.H.; Fisher, G.; Rahma, O.; Lyman, J.P.; et al. Sotigalimab and/or nivolumab with chemotherapy in first-line metastatic pancreatic cancer: Clinical and immunologic analyses from the randomized phase 2 PRINCE trial. Nat. Med. 2022, 28, 1167–1177. [Google Scholar] [CrossRef]
  71. Renouf, D.J.; Loree, J.M.; Knox, J.J.; Topham, J.T.; Kavan, P.; Jonker, D.; Welch, S.; Couture, F.; Lemay, F.; Tehfe, M.; et al. The CCTG PA.7 phase II trial of gemcitabine and nab-paclitaxel with or without durvalumab and tremelimumab as initial therapy in metastatic pancreatic ductal adenocarcinoma. Nat. Commun. 2022, 13, 5020. [Google Scholar] [CrossRef] [PubMed]
  72. Dean, A.; Gill, S.; McGregor, M.; Broadbridge, V.; Järveläinen, H.A.; Price, T. Dual αV-integrin and neuropilin-1 targeting peptide CEND-1 plus nab-paclitaxel and gemcitabine for the treatment of metastatic pancreatic ductal adenocarcinoma: A first-in-human, open-label, multicentre, phase 1 study. Lancet Gastroenterol. Hepatol. 2022, 7, 943–951. [Google Scholar] [CrossRef] [PubMed]
  73. Ikenaga, N.; Miyasaka, Y.; Ohtsuka, T.; Nakata, K.; Adachi, T.; Eguchi, S.; Nishihara, K.; Inomata, M.; Kurahara, H.; Hisaka, T.; et al. A Prospective Multicenter Phase II Trial of Neoadjuvant Chemotherapy with Gemcitabine Plus Nab-Paclitaxel for Borderline Resectable Pancreatic Cancer with Arterial Involvement. Ann. Surg. Oncol. 2023, 30, 193–202. [Google Scholar] [CrossRef]
  74. Grierson, P.M.; Tan, B.; Pedersen, K.S.; Park, H.; Suresh, R.; Amin, M.A.; Trikalinos, N.A.; Knoerzer, D.; Kreider, B.; Reddy, A.; et al. Phase Ib Study of Ulixertinib Plus Gemcitabine and Nab-Paclitaxel in Patients with Metastatic Pancreatic Adenocarcinoma. Oncologist 2023, 28, e115–e123. [Google Scholar] [CrossRef] [PubMed]
  75. Ozaka, M.; Nakachi, K.; Kobayashi, S.; Ohba, A.; Imaoka, H.; Terashima, T.; Ishii, H.; Mizusawa, J.; Katayama, H.; Kataoka, T.; et al. A randomised phase II study of modified FOLFIRINOX versus gemcitabine plus nab-paclitaxel for locally advanced pancreatic cancer (JCOG1407). Eur. J. Cancer 2023, 181, 135–144. [Google Scholar] [CrossRef] [PubMed]
  76. Feig, C.; Gopinathan, A.; Neesse, A.; Chan, D.S.; Cook, N.; Tuveson, D.A. The pancreas cancer microenvironment. Clin. Cancer Res. 2012, 18, 4266–4276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Mantoni, T.S.; Schendel, R.R.; Rödel, F.; Niedobitek, G.; Al-Assar, O.; Masamune, A.; Brunner, T.B. Stromal SPARC expression and patient survival after chemoradiation for non-resectable pancreatic adenocarcinoma. Cancer Biol. Ther. 2008, 7, 1806–1815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Provenzano, P.P.; Cuevas, C.; Chang, A.E.; Goel, V.K.; Von Hoff, D.D.; Hingorani, S.R. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012, 21, 418–429. [Google Scholar] [CrossRef] [Green Version]
  79. Jacobetz, M.A.; Chan, D.S.; Neesse, A.; Bapiro, T.E.; Cook, N.; Frese, K.K.; Feig, C.; Nakagawa, T.; Caldwell, M.E.; Zecchini, H.I.; et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013, 62, 112–120. [Google Scholar] [CrossRef] [Green Version]
  80. Du, J.; Gu, J.; Li, J. Mechanisms of drug resistance of pancreatic ductal adenocarcinoma at different levels. Biosci. Rep. 2020, 40. [Google Scholar] [CrossRef]
  81. Erkan, M.; Kurtoglu, M.; Kleeff, J. The role of hypoxia in pancreatic cancer: A potential therapeutic target? Expert. Rev. Gastroenterol. Hepatol. 2016, 10, 301–316. [Google Scholar] [CrossRef]
  82. Sparano, J.A. Defining a role and predicting benefit from platinum-based therapy in breast cancer: An evolving story. J. Clin. Oncol. 2015, 33, 1–3. [Google Scholar] [CrossRef]
  83. Catenacci, D.V.; Junttila, M.R.; Karrison, T.; Bahary, N.; Horiba, M.N.; Nattam, S.R.; Marsh, R.; Wallace, J.; Kozloff, M.; Rajdev, L.; et al. Randomized Phase Ib/II Study of Gemcitabine Plus Placebo or Vismodegib, a Hedgehog Pathway Inhibitor, in Patients With Metastatic Pancreatic Cancer. J. Clin. Oncol. 2015, 33, 4284–4292. [Google Scholar] [CrossRef]
  84. Chan, J.S.K.; Sng, M.K.; Teo, Z.Q.; Chong, H.C.; Twang, J.S.; Tan, N.S. Targeting nuclear receptors in cancer-associated fibroblasts as concurrent therapy to inhibit development of chemoresistant tumors. Oncogene 2018, 37, 160–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Yang, Z.; Zhao, N.; Cui, J.; Wu, H.; Xiong, J.; Peng, T. Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cell Oncol. 2020, 43, 123–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Mackey, J.R.; Mani, R.S.; Selner, M.; Mowles, D.; Young, J.D.; Belt, J.A.; Crawford, C.R.; Cass, C.E. Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res. 1998, 58, 4349–4357. [Google Scholar]
  87. Amrutkar, M.; Gladhaug, I.P. Pancreatic Cancer Chemoresistance to Gemcitabine. Cancers 2017, 9, 157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Arumugam, T.; Ramachandran, V.; Fournier, K.F.; Wang, H.; Marquis, L.; Abbruzzese, J.L.; Gallick, G.E.; Logsdon, C.D.; McConkey, D.J.; Choi, W. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009, 69, 5820–5828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Maloney, S.M.; Hoover, C.A.; Morejon-Lasso, L.V.; Prosperi, J.R. Mechanisms of Taxane Resistance. Cancers 2020, 12, 3323. [Google Scholar] [CrossRef]
  90. El Hassouni, B.; Li Petri, G.; Liu, D.S.K.; Cascioferro, S.; Parrino, B.; Hassan, W.; Diana, P.; Ali, A.; Frampton, A.E.; Giovannetti, E. Pharmacogenetics of treatments for pancreatic cancer. Expert. Opin. Drug Metab. Toxicol. 2019, 15, 437–447. [Google Scholar] [CrossRef]
  91. McCarroll, J.A.; Sharbeen, G.; Liu, J.; Youkhana, J.; Goldstein, D.; McCarthy, N.; Limbri, L.F.; Dischl, D.; Ceyhan, G.O.; Erkan, M.; et al. βIII-tubulin: A novel mediator of chemoresistance and metastases in pancreatic cancer. Oncotarget 2015, 6, 2235–2249. [Google Scholar] [CrossRef] [Green Version]
  92. Guo, F.; Li, J.; Qi, Y.; Hou, J.; Chen, H.; Jiang, S.W. HE4 overexpression decreases pancreatic cancer Capan-1 cell sensitivity to paclitaxel via cell cycle regulation. Cancer Cell Int. 2020, 20, 163. [Google Scholar] [CrossRef]
  93. Parasido, E.; Avetian, G.S.; Naeem, A.; Graham, G.; Pishvaian, M.; Glasgow, E.; Mudambi, S.; Lee, Y.; Ihemelandu, C.; Choudhry, M.; et al. The Sustained Induction of c-MYC Drives Nab-Paclitaxel Resistance in Primary Pancreatic Ductal Carcinoma Cells. Mol. Cancer Res. 2019, 17, 1815–1827. [Google Scholar] [CrossRef]
  94. Comandatore, A.; Immordino, B.; Balsano, R.; Capula, M.; Garajovà, I.; Ciccolini, J.; Giovannetti, E.; Morelli, L. Potential Role of Exosomes in the Chemoresistance to Gemcitabine and Nab-Paclitaxel in Pancreatic Cancer. Diagnostics 2022, 12. [Google Scholar] [CrossRef] [PubMed]
  95. Pandilla, R.; Kotapalli, V.; Gowrishankar, S.; Chigurupati, M.; Patnaik, S.; Uppin, S.; Rao, S.; Kalidindi, N.; Regulagadda, S.; Sundaram, C.; et al. Distinct genetic aberrations in oesophageal adeno and squamous carcinoma. Eur. J. Clin. Investig. 2013, 43, 1233–1239. [Google Scholar] [CrossRef] [PubMed]
  96. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  97. Hassan, M.S.; Awasthi, N.; Li, J.; Williams, F.; Schwarz, M.A.; Schwarz, R.E.; von Holzen, U. Superior Therapeutic Efficacy of Nanoparticle Albumin Bound Paclitaxel Over Cremophor-Bound Paclitaxel in Experimental Esophageal Adenocarcinoma. Transl. Oncol. 2018, 11, 426–435. [Google Scholar] [CrossRef]
  98. Wang, H.Y.; Yao, Z.H.; Tang, H.; Zhao, Y.; Zhang, X.S.; Yao, S.N.; Yang, S.J.; Liu, Y.Y. Weekly nanoparticle albumin-bound paclitaxel in combination with cisplatin versus weekly solvent-based paclitaxel plus cisplatin as first-line therapy in Chinese patients with advanced esophageal squamous cell carcinoma. Onco-Targets Ther. 2016, 9, 5663–5669. [Google Scholar] [CrossRef] [Green Version]
  99. Shi, Y.; Qin, R.; Wang, Z.K.; Dai, G.H. Nanoparticle albumin-bound paclitaxel combined with cisplatin as the first-line treatment for metastatic esophageal squamous cell carcinoma. Onco-Targets Ther. 2013, 6, 585–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Jiang, C.; Liao, F.X.; Rong, Y.M.; Yang, Q.; Yin, C.X.; He, W.Z.; Cai, X.Y.; Guo, G.F.; Qiu, H.J.; Chen, X.X.; et al. Efficacy of taxane-based regimens in a first-line setting for recurrent and/or metastatic Chinese patients with esophageal cancer. Asian Pac. J. Cancer Prev. 2014, 15, 5493–5498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Wang, D.; Zhang, W.; Qian, D.; Guan, Y.; Chen, X.; Zhang, H.; Wang, J.; Pang, Q. Efficacy and safety of weekly nab-paclitaxel plus cisplatin with concurrent intensity-modulated radiotherapy in patients with inoperable, locally advanced esophageal cancer: A pilot trial. Onco Targets Ther. 2018, 11, 6333–6338. [Google Scholar] [CrossRef] [Green Version]
  102. Huang, R.; Qiu, Z.; Zheng, C.; Zeng, R.; Chen, W.; Wang, S.; Li, E.; Xu, Y. Neoadjuvant Therapy for Locally Advanced Esophageal Cancers. Front. Oncol. 2022, 12, 734581. [Google Scholar] [CrossRef]
  103. Fan, Y.; Jiang, Y.; Zhou, X.; Chen, Q.; Huang, Z.; Xu, Y.; Gong, L.; Yu, H.; Yang, H.; Liu, J.; et al. Phase II study of neoadjuvant therapy with nab-paclitaxel and cisplatin followed by surgery in patients with locally advanced esophageal squamous cell carcinoma. Oncotarget 2016, 7, 50624–50634. [Google Scholar] [CrossRef] [Green Version]
  104. Tang, H.R.; Ma, H.F.; An, S.M.; Badakhshi, H.; Deng, J.Y.; Zhang, J.H.; Chen, Y.; Zhang, Z.; Guo, X.M.; Jiang, G.L.; et al. A Phase II Study of Concurrent Chemoradiotherapy With Paclitaxel and Cisplatin for Inoperable Esophageal Squamous Cell Carcinoma. Am. J. Clin. Oncol. 2016, 39, 350–354. [Google Scholar] [CrossRef]
  105. Yang, G.; Su, X.; Yang, H.; Luo, G.; Gao, C.; Zheng, Y.; Xie, W.; Huang, M.; Bei, T.; Bai, Y.; et al. Neoadjuvant programmed death-1 blockade plus chemotherapy in locally advanced esophageal squamous cell carcinoma. Ann. Transl. Med. 2021, 9, 1254. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, J.; Yang, Y.; Liu, Z.; Fu, X.; Cai, X.; Li, H.; Zhu, L.; Shen, Y.; Zhang, H.; Sun, Y.; et al. Multicenter, single-arm, phase II trial of camrelizumab and chemotherapy as neoadjuvant treatment for locally advanced esophageal squamous cell carcinoma. J. Immunother. Cancer 2022, 10. [Google Scholar] [CrossRef] [PubMed]
  107. Yan, X.; Duan, H.; Ni, Y.; Zhou, Y.; Wang, X.; Qi, H.; Gong, L.; Liu, H.; Tian, F.; Lu, Q.; et al. Tislelizumab combined with chemotherapy as neoadjuvant therapy for surgically resectable esophageal cancer: A prospective, single-arm, phase II study (TD-NICE). Int. J. Surg. 2022, 103, 106680. [Google Scholar] [CrossRef] [PubMed]
  108. Liu, J.; Li, J.; Lin, W.; Shao, D.; Depypere, L.; Zhang, Z.; Li, Z.; Cui, F.; Du, Z.; Zeng, Y.; et al. Neoadjuvant camrelizumab plus chemotherapy for resectable, locally advanced esophageal squamous cell carcinoma (NIC-ESCC2019): A multicenter, phase 2 study. Int. J. Cancer 2022, 151, 128–137. [Google Scholar] [CrossRef]
  109. Yang, Y.; Li, H.; Chen, X.; Qin, J.; Li, Y.; Shen, Y.; Zhang, R.; Kang, X.; Wang, Z.; Zheng, Q.; et al. Comparison of neoadjuvant nab-paclitaxel plus immunotherapy versus paclitaxel plus immunotherapy for esophageal squamous cell carcinoma. Thorac. Cancer 2023. [Google Scholar] [CrossRef]
  110. Yang, W.; Xing, X.; Yeung, S.J.; Wang, S.; Chen, W.; Bao, Y.; Wang, F.; Feng, S.; Peng, F.; Wang, X.; et al. Neoadjuvant programmed cell death 1 blockade combined with chemotherapy for resectable esophageal squamous cell carcinoma. J. Immunother. Cancer 2022, 10. [Google Scholar] [CrossRef]
  111. Sasaki, Y.; Nishina, T.; Yasui, H.; Goto, M.; Muro, K.; Tsuji, A.; Koizumi, W.; Toh, Y.; Hara, T.; Miyata, Y. Phase II trial of nanoparticle albumin-bound paclitaxel as second-line chemotherapy for unresectable or recurrent gastric cancer. Cancer Sci. 2014, 105, 812–817. [Google Scholar] [CrossRef] [Green Version]
  112. Bando, H.; Shimodaira, H.; Fujitani, K.; Takashima, A.; Yamaguchi, K.; Nakayama, N.; Takahashi, T.; Oki, E.; Azuma, M.; Nishina, T.; et al. A phase II study of nab-paclitaxel in combination with ramucirumab in patients with previously treated advanced gastric cancer. Eur. J. Cancer 2018, 91, 86–91. [Google Scholar] [CrossRef] [Green Version]
  113. Sato, S.; Kunisaki, C.; Tanaka, Y.; Sato, K.; Miyamoto, H.; Yukawa, N.; Fujii, Y.; Kimura, J.; Takagawa, R.; Takahashi, M.; et al. A Phase II Study of Tri-weekly Low-dose Nab-paclitaxel Chemotherapy for Patients with Advanced Gastric Cancer. Anticancer Res. 2018, 38, 6911–6917. [Google Scholar] [CrossRef]
  114. Takashima, A.; Shitara, K.; Fujitani, K.; Koeda, K.; Hara, H.; Nakayama, N.; Hironaka, S.; Nishikawa, K.; Kimura, Y.; Amagai, K.; et al. Peritoneal metastasis as a predictive factor for nab-paclitaxel in patients with pretreated advanced gastric cancer: An exploratory analysis of the phase III ABSOLUTE trial. Gastric Cancer 2019, 22, 155–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kobayashi, D.; Mochizuki, Y.; Torii, K.; Takeda, S.; Kawase, Y.; Ishigure, K.; Teramoto, H.; Ando, M.; Kodera, Y. Phase II multi-institutional prospective trial of nab-paclitaxel as second-line chemotherapy for advanced gastric cancer refractory to fluoropyrimidine with modified dose reduction criteria (CCOG1303). Int. J. Clin. Oncol. 2020, 25, 1793–1799. [Google Scholar] [CrossRef] [PubMed]
  116. Tamura, S.; Taniguchi, H.; Nishikawa, K.; Imamura, H.; Fujita, J.; Takeno, A.; Matsuyama, J.; Kimura, Y.; Kawada, J.; Hirao, M.; et al. A phase II trial of dose-reduced nab-paclitaxel for patients with previously treated, advanced or recurrent gastric cancer (OGSG 1302). Int. J. Clin. Oncol. 2020, 25, 2035–2043. [Google Scholar] [CrossRef]
  117. Sahai, V.; Catalano, P.J.; Zalupski, M.M.; Lubner, S.J.; Menge, M.R.; Nimeiri, H.S.; Munshi, H.G.; Benson, A.B., 3rd; O’Dwyer, P.J. Nab-Paclitaxel and Gemcitabine as First-line Treatment of Advanced or Metastatic Cholangiocarcinoma: A Phase 2 Clinical Trial. JAMA Oncol. 2018, 4, 1707–1712. [Google Scholar] [CrossRef]
  118. Shroff, R.T.; Javle, M.M.; Xiao, L.; Kaseb, A.O.; Varadhachary, G.R.; Wolff, R.A.; Raghav, K.P.S.; Iwasaki, M.; Masci, P.; Ramanathan, R.K.; et al. Gemcitabine, Cisplatin, and nab-Paclitaxel for the Treatment of Advanced Biliary Tract Cancers: A Phase 2 Clinical Trial. JAMA Oncol. 2019, 5, 824–830. [Google Scholar] [CrossRef] [PubMed]
  119. Ducreux, M.; Bennouna, J.; Adenis, A.; Conroy, T.; Lièvre, A.; Portales, F.; Jeanes, J.; Li, L.; Romano, A. Efficacy and safety of nab-paclitaxel in patients with previously treated metastatic colorectal cancer: A phase II COLO-001 trial. Cancer Chemother. Pharmacol. 2017, 79, 9–16. [Google Scholar] [CrossRef] [PubMed]
  120. Overman, M.J.; Adam, L.; Raghav, K.; Wang, J.; Kee, B.; Fogelman, D.; Eng, C.; Vilar, E.; Shroff, R.; Dasari, A.; et al. Phase II study of nab-paclitaxel in refractory small bowel adenocarcinoma and CpG island methylator phenotype (CIMP)-high colorectal cancer. Ann. Oncol. 2018, 29, 139–144. [Google Scholar] [CrossRef] [PubMed]
  121. Wagner, A.D.; Syn, N.L.; Moehler, M.; Grothe, W.; Yong, W.P.; Tai, B.C.; Ho, J.; Unverzagt, S. Chemotherapy for advanced gastric cancer. Cochrane Database Syst. Rev. 2017, 8, Cd004064. [Google Scholar] [CrossRef] [Green Version]
  122. Park, S.R.; Chun, J.H.; Kim, Y.W.; Lee, J.H.; Choi, I.J.; Kim, C.G.; Lee, J.S.; Bae, J.M.; Kim, H.K. Phase II study of low-dose docetaxel/fluorouracil/cisplatin in metastatic gastric carcinoma. Am. J. Clin. Oncol. 2005, 28, 433–438. [Google Scholar] [CrossRef]
  123. Sun, D.S.; Jeon, E.K.; Won, H.S.; Park, J.C.; Shim, B.Y.; Park, S.Y.; Hong, Y.S.; Kim, H.K.; Ko, Y.H. Outcomes in elderly patients treated with a single-agent or combination regimen as first-line chemotherapy for recurrent or metastatic gastric cancer. Gastric Cancer 2015, 18, 644–652. [Google Scholar] [CrossRef]
  124. Narahara, H.; Iishi, H.; Imamura, H.; Tsuburaya, A.; Chin, K.; Imamoto, H.; Esaki, T.; Furukawa, H.; Hamada, C.; Sakata, Y. Randomized phase III study comparing the efficacy and safety of irinotecan plus S-1 with S-1 alone as first-line treatment for advanced gastric cancer (study GC0301/TOP-002). Gastric Cancer 2011, 14, 72–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Al-Batran, S.E.; Hofheinz, R.D.; Pauligk, C.; Kopp, H.G.; Haag, G.M.; Luley, K.B.; Meiler, J.; Homann, N.; Lorenzen, S.; Schmalenberg, H.; et al. Histopathological regression after neoadjuvant docetaxel, oxaliplatin, fluorouracil, and leucovorin versus epirubicin, cisplatin, and fluorouracil or capecitabine in patients with resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4-AIO): Results from the phase 2 part of a multicentre, open-label, randomised phase 2/3 trial. Lancet Oncol. 2016, 17, 1697–1708. [Google Scholar] [CrossRef] [PubMed]
  126. Kim, H.S.; Kim, H.J.; Kim, S.Y.; Kim, T.Y.; Lee, K.W.; Baek, S.K.; Kim, T.Y.; Ryu, M.H.; Nam, B.H.; Zang, D.Y. Second-line chemotherapy versus supportive cancer treatment in advanced gastric cancer: A meta-analysis. Ann. Oncol. 2013, 24, 2850–2854. [Google Scholar] [CrossRef] [PubMed]
  127. Iacovelli, R.; Pietrantonio, F.; Farcomeni, A.; Maggi, C.; Palazzo, A.; Ricchini, F.; de Braud, F.; Di Bartolomeo, M. Chemotherapy or targeted therapy as second-line treatment of advanced gastric cancer. A systematic review and meta-analysis of published studies. PLoS ONE 2014, 9, e108940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ghosn, M.; Tabchi, S.; Kourie, H.R.; Tehfe, M. Metastatic gastric cancer treatment: Second line and beyond. World J. Gastroenterol. 2016, 22, 3069–3077. [Google Scholar] [CrossRef]
  129. Sym, S.J.; Hong, J.; Park, J.; Cho, E.K.; Lee, J.H.; Park, Y.H.; Lee, W.K.; Chung, M.; Kim, H.S.; Park, S.H.; et al. A randomized phase II study of biweekly irinotecan monotherapy or a combination of irinotecan plus 5-fluorouracil/leucovorin (mFOLFIRI) in patients with metastatic gastric adenocarcinoma refractory to or progressive after first-line chemotherapy. Cancer Chemother. Pharmacol. 2013, 71, 481–488. [Google Scholar] [CrossRef]
  130. Ford, H.E.; Marshall, A.; Bridgewater, J.A.; Janowitz, T.; Coxon, F.Y.; Wadsley, J.; Mansoor, W.; Fyfe, D.; Madhusudan, S.; Middleton, G.W.; et al. Docetaxel versus active symptom control for refractory oesophagogastric adenocarcinoma (COUGAR-02): An open-label, phase 3 randomised controlled trial. Lancet Oncol. 2014, 15, 78–86. [Google Scholar] [CrossRef]
  131. Awasthi, N.; Schwarz, M.A.; Zhang, C.; Klinz, S.G.; Meyer-Losic, F.; Beaufils, B.; Thiagalingam, A.; Schwarz, R.E. Augmenting Experimental Gastric Cancer Activity of Irinotecan through Liposomal Formulation and Antiangiogenic Combination Therapy. Mol. Cancer Ther. 2022, 21, 1149–1159. [Google Scholar] [CrossRef]
  132. Grojean, M.; Schwarz, M.A.; Schwarz, J.R.; Hassan, S.; von Holzen, U.; Zhang, C.; Schwarz, R.E.; Awasthi, N. Targeted dual inhibition of c-Met/VEGFR2 signalling by foretinib improves antitumour effects of nanoparticle paclitaxel in gastric cancer models. J. Cell Mol. Med. 2021, 25, 4950–4961. [Google Scholar] [CrossRef]
  133. Crawford, K.; Bontrager, E.; Schwarz, M.A.; Chaturvedi, A.; Lee, D.D.; Md Sazzad, H.; von Holzen, U.; Zhang, C.; Schwarz, R.E.; Awasthi, N. Targeted FGFR/VEGFR/PDGFR inhibition with dovitinib enhances the effects of nab-paclitaxel in preclinical gastric cancer models. Cancer Biol. Ther. 2021, 22, 619–629. [Google Scholar] [CrossRef]
  134. Awasthi, N.; Schwarz, M.A.; Zhang, C.; Schwarz, R.E. Augmentation of Nab-Paclitaxel Chemotherapy Response by Mechanistically Diverse Antiangiogenic Agents in Preclinical Gastric Cancer Models. Mol. Cancer Ther. 2018, 17, 2353–2364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Zhang, C.; Awasthi, N.; Schwarz, M.A.; Schwarz, R.E. The dual PI3K/mTOR inhibitor NVP-BEZ235 enhances nab-paclitaxel antitumor response in experimental gastric cancer. Int. J. Oncol. 2013, 43, 1627–1635. [Google Scholar] [CrossRef] [Green Version]
  136. Shi, W.J.; Gao, J.B. Molecular mechanisms of chemoresistance in gastric cancer. World J. Gastrointest. Oncol. 2016, 8, 673–681. [Google Scholar] [CrossRef] [PubMed]
  137. Watson, S.; de la Fouchardière, C.; Kim, S.; Cohen, R.; Bachet, J.B.; Tournigand, C.; Ferraz, J.M.; Lefevre, M.; Colin, D.; Svrcek, M.; et al. Oxaliplatin, 5-Fluorouracil and Nab-paclitaxel as perioperative regimen in patients with resectable gastric adenocarcinoma: A GERCOR phase II study (FOXAGAST). Eur. J. Cancer 2019, 107, 46–52. [Google Scholar] [CrossRef]
  138. Blechacz, B.; Gores, G.J. Cholangiocarcinoma: Advances in pathogenesis, diagnosis, and treatment. Hepatology 2008, 48, 308–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Macias, R.I. Cholangiocarcinoma: Biology, Clinical Management, and Pharmacological Perspectives. ISRN Hepatol. 2014, 2014, 828074. [Google Scholar] [CrossRef]
  140. Rizvi, S.; Gores, G.J. Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 2013, 145, 1215–1229. [Google Scholar] [CrossRef] [Green Version]
  141. Gusani, N.J.; Balaa, F.K.; Steel, J.L.; Geller, D.A.; Marsh, J.W.; Zajko, A.B.; Carr, B.I.; Gamblin, T.C. Treatment of unresectable cholangiocarcinoma with gemcitabine-based transcatheter arterial chemoembolization (TACE): A single-institution experience. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract. 2008, 12, 129–137. [Google Scholar] [CrossRef]
  142. Valle, J.; Wasan, H.; Palmer, D.H.; Cunningham, D.; Anthoney, A.; Maraveyas, A.; Madhusudan, S.; Iveson, T.; Hughes, S.; Pereira, S.P.; et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N. Engl. J. Med. 2010, 362, 1273–1281. [Google Scholar] [CrossRef] [Green Version]
  143. Woodford, R.; Brungs, D.; Leighton, C.; Grimison, P.; Sjoquist, K.M.; Becker, T.; Robinson, S.; Gebski, V.; Wilson, K.; Chantrill, L.; et al. Combination chemotherapy with NAB(®) -paclitaxel and capecitabine for patients with advanced biliary tract cancer (NAP-CAPABIL Pilot Study). Asia Pac. J. Clin. Oncol. 2022, 18, e220–e226. [Google Scholar] [CrossRef]
  144. Rawla, P.; Sunkara, T.; Barsouk, A. Epidemiology of colorectal cancer: Incidence, mortality, survival, and risk factors. Prz. Gastroenterol. 2019, 14, 89–103. [Google Scholar] [CrossRef] [PubMed]
  145. Einzig, A.I.; Neuberg, D.; Wiernik, P.H.; Grochow, L.B.; Ramirez, G.; O’Dwyer, P.J.; Petrelli, N.J. Phase II Trial of Paclitaxel in Patients with Advanced Colon Cancer Previously Untreated with Cytotoxic Chemotherapy: An Eastern Cooperative Oncology Group Trial (PA286). Am. J. Ther. 1996, 3, 750–754. [Google Scholar] [CrossRef] [PubMed]
  146. Yusuf, R.Z.; Duan, Z.; Lamendola, D.E.; Penson, R.T.; Seiden, M.V. Paclitaxel resistance: Molecular mechanisms and pharmacologic manipulation. Curr. Cancer Drug Targets 2003, 3, 1–19. [Google Scholar] [CrossRef]
  147. Cordon-Cardo, C.; O’Brien, J.P.; Boccia, J.; Casals, D.; Bertino, J.R.; Melamed, M.R. Expression of the multidrug resistance gene product (P-glycoprotein) in human normal and tumor tissues. J. Histochem. Cytochem. 1990, 38, 1277–1287. [Google Scholar] [CrossRef]
  148. Jang, S.H.; Wientjes, M.G.; Au, J.L. Kinetics of P-glycoprotein-mediated efflux of paclitaxel. J. Pharmacol. Exp. Ther. 2001, 298, 1236–1242. [Google Scholar] [PubMed]
  149. Koziara, J.M.; Whisman, T.R.; Tseng, M.T.; Mumper, R.J. In-vivo efficacy of novel paclitaxel nanoparticles in paclitaxel-resistant human colorectal tumors. J. Control Release 2006, 112, 312–319. [Google Scholar] [CrossRef] [PubMed]
  150. Swanton, C.; Tomlinson, I.; Downward, J. Chromosomal instability, colorectal cancer and taxane resistance. Cell Cycle 2006, 5, 818–823. [Google Scholar] [CrossRef] [Green Version]
  151. Hartley, C.; Rowan, D.; Chen, X.; Gomez-Arellano, L.; West, A.M.; Oshima, K.; Mackinnon, A.C. Increased SPARC expression is associated with neoadjuvant therapy in resectable pancreatic ductal adenocarcinoma. Pract. Lab. Med. 2020, 21, e00171. [Google Scholar] [CrossRef]
  152. Hidalgo, M.; Plaza, C.; Musteanu, M.; Illei, P.; Brachmann, C.B.; Heise, C.; Pierce, D.; Lopez-Casas, P.P.; Menendez, C.; Tabernero, J.; et al. SPARC Expression Did Not Predict Efficacy of nab-Paclitaxel plus Gemcitabine or Gemcitabine Alone for Metastatic Pancreatic Cancer in an Exploratory Analysis of the Phase III MPACT Trial. Clin. Cancer Res. 2015, 21, 4811–4818. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Molecular Structure of nab-paclitaxel.
Figure 1. Molecular Structure of nab-paclitaxel.
Biomedicines 11 02000 g001
Figure 2. Molecular site of action of nab-paclitaxel.
Figure 2. Molecular site of action of nab-paclitaxel.
Biomedicines 11 02000 g002
Figure 3. Schematic diagram of mechanisms of action of nab-paclitaxel and conventional paclitaxel.
Figure 3. Schematic diagram of mechanisms of action of nab-paclitaxel and conventional paclitaxel.
Biomedicines 11 02000 g003
Table 1. Phase II and III clinical trials of nab-paclitaxel in PDAC.
Table 1. Phase II and III clinical trials of nab-paclitaxel in PDAC.
Authors,PatientsTherapeuticORRMedian MedianCommon
Year Regimen(%)PFS (Mo) OS (Mo)Adverse Events %
Hematologic (Grade ≥ 3)
Von Hoff et al.,
2011 [39]
Untreated
advanced
NG487.912.2neutropenia 67
leukopenia 44
Thrombocytopenia 23
Hosein et al.,
2013 [40]
Gemcitabine
refractory
nab-pac581.77.3neutropenia 32
anemia 11
Von Hoff et al.,
2013 [41]
Untreated
metastatic
NG vs. gemcitabine23 vs. 75.5 vs. 3.78.5 vs. 6.7neutropenia 38 vs. 27
leukopenia 31 vs. 16
thrombocytopenia 13 vs. 9
anemia 13 vs. 12
Bachet et al.,
2017 [42].
untreated
metastatic
NG vs
nab-pac+
leucovorin/5-FU
at 4 m PFS rate
54% vs. 56%
neutropenia 32 vs. 23
thrombocytopenia 18 (NG
anemia 13 (NG)
Ko et al.,
2017 [43].
Untreated
metastatic
NG+apatorsen
vs NG
18
for both
2.7 vs. 3.85.3 vs. 6.9leukopenia 0 (both)
thrombocytopenia 0 (both)
Weiss et al.,
2018 [44].
untreated metastatic NG+
pembrolizumab
9.115neutropenia 47
thrombocytopenia 20
Hingorani et al.,
2018 [45]
untreated
metastatic
NG+PEGPH20
vs NG
In high hyaluronan patientsneutropenia 29 vs. 18
45 vs. 31 9.2 vs. 5.211.5 vs. 8.5thrombocytopenia 16 vs. 9
Cutsem et al.,
2020 [46].
untreated
Metastatic
High hyaluronan
NG+PEGPH20 vs. NG47 vs. 367.1 vs. 7.111.2 vs. 11.5thrombocytopenia 21 vs. 16
Zhang et al.,
2018 [47].
untreated
advanced
nab-pac+ S1
then S1 maintenance
53.16.213.6neutropenia 27.6
Reni et al.,
2018 [48].
untreated
metastatic
NG+cisplatin+
capecitabine vs. NG
at 6 months DFR
74 vs. 46%
neutropenia 41 vs. 39
anemia 21 vs. 22
Karasic et al.,
2019 [50].
untreated
advanced
NG+HCQ
vs NG
38.2 vs. 215.7 vs. 6.411.1 vs. 12.1neutropenia 42.6 vs. 22.6
anemia 3.7 vs. 17
Zeh et al.,
2020 [51].
Untreated
resectable
preoperative NG+
HCQ vs. NG
recurrence-free
survival
16.6 vs. 13.5
36 vs. 32all grade ≥ 3
adverse events
62.5 vs. 60.5
Hu et al.,
2019 [52].
Untreated
metastatic
NG+
Tarextumab
vs NG
20.2 vs. 31.83.7 vs. 5.56.4 vs. 7.9
neutropenia 9 vs. 18
thrombocytopenia 49 vs. 25
anemia 29 vs. 26
Kundranda
et al, 2020 [53].
untreated metastaticNG+
Istiratumab
vs NG
39.5 vs. 51.2high IGF-1
3.6 vs. 7.3
high IGF-1/HRG+
4.1 vs. 7.3
8.9 vs. 11.7neutropenia 30 vs. 34
anemia 18.6 vs. 18.2
thrombocytopenia 16 vs. 7
De Jesus-Acosta
et al, 2020 [54].
untreated
metastatic
NG+
vismodegib
405.49.8No data about
neutropenia, anemia or thrombocytopenia
O’Reilly et al.,
2020 [55].
Untreated
metastatic
NG+
Necuparanib
vs NG
5.5 vs. 6.910.7 vs. 9.99neutropenia 33 vs. 34
thrombocytopenia 27 vs. 5
anemia 22 vs. 11
Barbour et al.,
2020 [56].
resectable Perioperative
NG
12.323.5neutropenia 40
anemia 10
thrombocytopenia 5
Raufi et al.,
2020 [58].
resected adjuvant FOLFOX
plus nab-pac
19.753.5neutropenia 26
Kunzmann
et al, 2021 [64].
locally advanced
resectable
NG vs. NG followed
by FOLFIRINOX
surgical conversion rate
35.9% vs. 43.9%
18.5 vs. 20.7neutropenia 28 vs. 24
Tempero
et al, 2021 [65].
untreated
metastatic
NG+ibrutinib vs. NG29 vs. 425.3 vs. 6 9.7 vs. 10.8 neutropenia 24 vs. 35
anemia 16 vs. 17
Giommoni et al., 2021 [66].untreated metastaticnab-pac+FOLFIRI
vs nab-pac+FOLFOX
31 for both6 vs. 5.6 10.2 vs. 10.4neutropenia 19 vs. 29
febrile neutropenia 12 vs. 1
thrombocytopenia 2.4 vs. 0
anemia 7 vs. 10
Kondo et al.,
2021 [67].
borderlineresectable
arterial contact
Neoadjuvant
NG+S-1
4324.241neutropenia 25
leukopenia 19
anemia 2
Hewitt
et al, 2022 [68].
Borderline
resectable or locally advancedunresectable
soc [(FOLFIRINOX or
NG) plus chemoradition]
plus algenpantucel
vs soc + placebo
12.4 vs. 13.4 14.3 vs. 14.9all grade ≥ 3
adverse events
81 vs. 75
Babiker et al.,
2022 [69].
untreated
metastatic
nab-pac
plus RX-3117
23.1 5.6 neutropenia 20
anemia 22
Padron et al.,
2022 [70].
untreated
metastatic
NG+nivolumabNG+sotigalimab NG+sotiga+nivo 50 vs. 33
vs 31
6.4 vs. 7.3 vs. 6.716.7 vs. 11.4 vs. 10.1 neutropenia 33, 54, 57
thrombocytopenia 11, 16, 60
anemia 33, 24, 51
Renouf et al.,
2022 [71].
untreated
metastatic
NG+ durvalumab
+tremelimumabvs NG
30.3 vs. 235.5 vs. 5.49.8 vs. 8.8 neutropenia 49 vs. 44
thrombocytopenia 11 vs. 16
anemia 22 vs. 32
lymphopenia 38 vs. 20
Ikenaga et al., 2023 [73]. borderline
resectable
with arterial
involvement
Neoadjuvant
NG
24.9neutropenia 68
leukopenia 39
thrombocytopenia 7
anemia 4
Ozaka et al.,
2023 [75].
untreated
locally
Advanced
mFOLFIRINOXvs NG30.9 vs. 42.111.2 vs. 9.423 vs. 21.3neutropenia 60 vs. 79
leukopenia 23 vs. 44
anemia 11 vs. 19
PDAC, pancreatic ductal adenocarcinoma; NG, nab-paclitaxel plus gemcitabine; ORR, overall response rate; PFS, progression-free survival; OS, overall survival; mo, months; vs, versus; nab-pac, nab-paclitaxel, 5-FU, 5-fluorouracil; PEGPH20, pegvorhyaluronidase alfa; HCQ, hydroxychloroquine; FOLFIRINOX, 5-fluorouracil/leucovorin plus irinotecan plus oxaliplatin; FOLFIRI, folinic acid plus 5-fluorouracil plus irinotecan; FOLFOX, folinic acid plus 5-fluorouracil plus oxaliplatin; soc, standard of care.
Table 2. Phase II and III clinical trials of nab-paclitaxel in esophageal, gastric, bile duct and colorectal cancers.
Table 2. Phase II and III clinical trials of nab-paclitaxel in esophageal, gastric, bile duct and colorectal cancers.
Authors,PatientsTherapeuticORRMedian Median Common
Year Regimen(%)PFS (Mo) OS (Mo)Adverse Events %
Hematologic (Grade ≥ 3)
Yun Fan et al., 2016 [103]. neoadjuvantlocally advancedESCC Nab-paclitaxel + cisplatin ORR 65.7% 34.7 37.8neutropenia 11.5
anemia 8.6
thrombocytopenia 5
Guozhen Yang
et al, 2021 [105].
neoadjuvant locally advancedESCCNab-paclitaxel + camrelizumab + S133.33% (cPR)no surgicaldelay no preoperativedeathneutropenia 0
anemia 0
thrombocytopenia 0
Jun Liuet al, 2022 [106]. neoadjuvantlocally advancedESCC Nab-paclitaxel+ carboplatin+ camrelizumab 39.2% (cPR) no surgicaldelayno preoperativedeath neutropenia 50
anemia 6.7 thrombocytopenia 6.7
Yafan Yang et al., 2023 [109]. neoadjuvant locally resectable
ESCCICIs+ paclitaxel
+ cisplatin
ICIs+ Nab-paclitaxel+ cisplatin vs. 36.7% (cPR) vs. 21.4% (cPR)PFS not yet reachedOS not yet reachednot yet reached
YasutsunaSasaki et al., 2014 [111].previously treated resectable or recurrent GCNab-paclitaxelORR 27.8% 2.9 9.2 neutropenia 49.1
anemia 7.3
thrombocytopenia 0
Hideaki Bonda et al., 2018 [112].
previouslytreated advanced GCNab-paclitaxel+ ramucirumab ORR 54.8% 7.6not yet reachedneutropenia 76.7
anemia 11.6
Sho Sato et al., 2018 [113].
previously treatedunresectable
or recurrent GC
Nab-paclitaxelORR 5.9% 2.49.2neutropenia 5.9
anemia 8.8
Atsuo Takashima et al., 2019 [114].
pretreated advancedPM GCNab-paclitaxel vs. sb-paclitaxel------- 4.0 vs. 2.67.6 vs. 4.9--------
DaisukeKobayashi et al., 2020 [115].previously treated advanced GC Nab-paclitaxel ORR 16% 3.5 9.0 neutropenia 49
anemia 2
thrombocytopenia 0
ShegeyukiTamura et al, 2020 [116]. previouslytreatedunresectable or recurrent GCNab-paclitaxel
(low dose)
ORR 3.1%2.2 6.3 neutropenia 37.5
anemia 12.5
thrombocytopenia 3.1
VaibhavSahaiet al, 2018 [117].First line therapyadvanced or metastatic CCANab-paclitaxel+ gemcitabineORR 30% 7.7 12.4neutropenia 43
anemia 15
thrombocytopenia 16
Rachna Shroff et al., 2019 [118]. First line therapy advanced BTCs Nab-paclitaxel + gemcitabine + cisplatin PRR 45% 11.819.2 neutropenia 33
anemia 16
thrombocytopenia 13
Michel Ducreux et al., 2017 [119].
pretreatedmCRCNab-paclitaxel ORR 0% 8.1 weeks --------neutropenia 22
anemia 5
Overman et al., 2018 [120].pretreated refractory SBAvsCIMP high CRCNab-paclitaxel ORR 20% vs. ORR 0% 3.2 vs. 2.1 not yet reachedneutropenia 9
anemia 0
thrombocytopenia 6
Abbreviations: ORR (overall response rate), cPR (complete pathological response), PFS (progressive free survival) OS (overall survival), ESCC (esophageal squamous cell carcinoma), GC (gastric cancer), PM GC (peritoneal metastasis gastric cancer), CCA (cholangiocarcinoma), BTCs (Biliary tract cancers), PRR (partial response rate), mCRC (metastatic colorectal cancer), SBA (small bowel adenocarcinoma), CIMP (CpG island methylator phenotype), CRC (colorectal cancer).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hassan, M.S.; Awasthi, N.; Ponna, S.; von Holzen, U. Nab-Paclitaxel in the Treatment of Gastrointestinal Cancers—Improvements in Clinical Efficacy and Safety. Biomedicines 2023, 11, 2000. https://doi.org/10.3390/biomedicines11072000

AMA Style

Hassan MS, Awasthi N, Ponna S, von Holzen U. Nab-Paclitaxel in the Treatment of Gastrointestinal Cancers—Improvements in Clinical Efficacy and Safety. Biomedicines. 2023; 11(7):2000. https://doi.org/10.3390/biomedicines11072000

Chicago/Turabian Style

Hassan, Md Sazzad, Niranjan Awasthi, Saisantosh Ponna, and Urs von Holzen. 2023. "Nab-Paclitaxel in the Treatment of Gastrointestinal Cancers—Improvements in Clinical Efficacy and Safety" Biomedicines 11, no. 7: 2000. https://doi.org/10.3390/biomedicines11072000

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop