You are on page 1of 8

Expert Review of Anticancer Therapy

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iery20

Emerging treatment options for prostate cancer

Mohammad Atiq, Elias Chandran, Fatima Karzai, Ravi A. Madan & Jeanny B.
Aragon-Ching

To cite this article: Mohammad Atiq, Elias Chandran, Fatima Karzai, Ravi A. Madan & Jeanny
B. Aragon-Ching (2023) Emerging treatment options for prostate cancer, Expert Review of
Anticancer Therapy, 23:6, 625-631, DOI: 10.1080/14737140.2023.2208352

To link to this article: https://doi.org/10.1080/14737140.2023.2208352

Published online: 02 May 2023.

Submit your article to this journal

Article views: 230

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iery20
EXPERT REVIEW OF ANTICANCER THERAPY
2023, VOL. 23, NO. 6, 625–631
https://doi.org/10.1080/14737140.2023.2208352

REVIEW

Emerging treatment options for prostate cancer


Mohammad Atiqa, Elias Chandran a
, Fatima Karzaia, Ravi A. Madana and Jeanny B. Aragon-Ching b,c

a
Genitourinary Malignancy Branch, National Cancer Institute, Bethesda, MD, USA; bGU Medical Oncology, Inova Schar Cancer Institute, Fairfax, VA,
USA; cMedical Education, University of Virginia, Charlottesville, VA, USA

ABSTRACT ARTICLE HISTORY


Introduction: Prostate cancer treatment has rapidly evolved in the past few years. Androgen depriva­ Received 4 January 2023
tion therapy has been the backbone of treatment for locally advanced and metastatic prostate cancer, Accepted 25 April 2023
but incremental benefits in survival have been shown by adding androgen-receptor pathway inhibitors KEYWORDS
(ARPI) across various spectrums of disease state. In addition, docetaxel chemotherapy remains the first- Anti-androgen;
line chemotherapy regimen available with survival benefits shown with triplet therapy in those who are immunotherapy; metastatic
chemotherapy eligible. However, disease progression remains inevitable and novel agents such as prostate cancer;
radioligand therapy with lutetium have shown improvement in survival. radioligands; prostate cancer
Areas covered: This review discusses the pivotal trials that led to the U.S. FDA approval of agents
utilized in metastatic prostate cancer and explores the use of novel agents including prostate-specific
membrane antigen-targeting agents, radioligands, cell-based therapy, chimeric antigen receptor T-
cell, BiTE, and antibody drug conjugates.
Expert opinion: Treatment landscape for metastatic castrate-resistant prostate cancer (mCRPC) has
evolved beyond additional agents with ARPI and/or docetaxel, including other treatments with sipu­
leucel-T, radium, cabazitaxel, PARP inhibitors, and lutetium, which have specific indications and roles in
sequencing. Novel therapies remain critically needed after progression from lutetium.

1. Introduction
cancer (mCRPC), docetaxel plus prednisone improved median
Prostate cancer remains the most common non-cutaneous overall survival (OS) compared to mitoxantrone plus predni­
malignancy among American men. It is estimated to occur in sone in the randomized phase III study TAX 327 (18.9 months
268,490 men and expected to result in 34,500 deaths in 2022 vs. 16.5 months) [5]. Docetaxel’s benefit when added to ADT in
[1]. A review of over 3 million prostate cancer diagnoses from metastatic castration-sensitive prostate cancer (mCSPC) was
2001 to 2017 revealed approximately 5% of new cases to be demonstrated in two studies: CHAARTED and an arm of the
metastatic at diagnosis with 5- and 10-year survivals of around multiplatform study, STAMPEDE [6,7]. The first of these studies
30% and 18%, respectively [2]. In addition, of patients defini­ yielded an improved OS of 57.6 months when docetaxel was
tively treated for localized prostate cancer, 20–30% will have added to ADT versus 44.0 months with ADT alone. Patients
recurrence in the form of rising prostate-specific antigen (PSA) with mCSPC treated in the ADT plus docetaxel arm of
or disseminated disease that necessitates further treatment STAMPEDE had a median OS of 81 months while those given
[3]. The original backbone for treatment of metastatic prostate ADT alone had a median OS of 71 months. Cabazitaxel only
cancer, androgen ablation or androgen deprivation therapy carries an approval in mCRPC. When used with prednisone in
(ADT), was discovered in the 1940s [4]. Since then, the treat­ mCRPC patients who had progressed on docetaxel, it had
ment landscape has evolved substantially. Many new FDA- a 2.4-month improvement in OS over mCRPC patients treated
approved therapies within the past decade have been with mitoxantrone plus prednisone [8]. It has also been shown
oriented around the androgen receptor; however, multiple to improve survival in patients with mCRPC post-docetaxel
novel avenues of treatment have emerged, demonstrating and have received either abiraterone or enzalutamide, com­
promise for expansion of the therapeutic tools available to pared to switching to the other of abiraterone or enzaluta­
treat this lethal disease. mide in the CARD trial: median OS 13.6 versus 11.0 months
(HR 0.64; 95% CI, 0.46–0.89) [9].
Androgen receptor pathway inhibitors (ARPI) with indica­
2. Current treatment landscape
tions for treatment of prostate cancer include the androgen
The current treatment landscape for metastatic prostate can­ synthesis inhibitor, abiraterone, and the three androgen
cer includes chemotherapy, anti-androgens, radiopharmaceu­ receptor inhibitors: enzalutamide, apalutamide, and darolu­
tical therapy, immunotherapy, and targeted therapies. The tamide. Abiraterone in combination with low-dose predni­
mainstay chemotherapies used in metastatic prostate cancer sone was first shown to improve OS in mCRPC post-
are docetaxel and cabazitaxel, both used in combination with docetaxel in the study COU-AA-301 [10,11]. Here, median
ADT. In patients with metastatic castration-resistant prostate OS was 15.8 months with abiraterone plus low-dose

CONTACT Jeanny B. Aragon-Ching jeanny.aragon-ching@inova.org GU Medical Oncology, Inova Schar Cancer Institute, Fairfax, VA, United States
This work was authored as part of the Contributor’s official duties as an Employee of the United States Government and is therefore a work of the United States Government. In accordance
with 17 USC 105, no copyright protection is available for such works under US Law.
626 M. ATIQ ET AL.

ADT in the PEACE-1 trial improved OS (HR 0.82, 95% CI,


Article highlights 0.69–0.98) [25].
● The treatment landscape for metastatic hormone-sensitive prostate
Radiopharmaceutical therapy has largely been composed
cancer has rapidly evolved with androgen deprivation therapy as of the alpha emitter Radium 223 dichloride. Radium 223
backbone and additional androgen-receptor pathway inhibitors dichloride was the first-in-class alpha-emitting particle that
(ARPI) or docetaxel or combination as triplet therapy as first-line
treatment.
showed improvement in OS in the treatment of mCRPC
● Treatment sequencing in metastatic castrate-resistant prostate cancer based on the ALSYMPCA trial [26]. Currently, Radium 223
(mCRPC) remains an important issue upon failure from hormone- dichloride is utilized in mCRPC patients with symptomatic
based therapies.
● Lutetium has recently been approved for mCRPC after failure from
bone metastases. Beta emitters have been used in mCRPC
one ARPI and docetaxel, with improvements seen in radiographic for many years in the form of samarium-153 and strontium-
progression-free survival and overall survival. 89. However, their role had only been in palliation for painful
● Emerging treatment options include radioactive ligand, antibody-
drug conjugates, chimeric antigen receptor T-cell, and cytokine-
bone metastases as neither provided survival benefit [27,28].
based therapies. The most recent advancement in radioligand therapy has
been with beta emitters, specifically lutetium-177 (177Lu),
which has been combined with prostate-specific membrane
antigen (PSMA), a type II transmembrane glycoprotein speci­
prednisone compared to 11.2 months for placebo. fically expressed in prostate cancer cells. TheraP, a phase II
A subsequent study of pre-docetaxel patients (COU-AA-302) study that compared 177Lu-PSMA-617 to cabazitaxel in
showed improved median OS favoring those treated with patients with mCRPC, showed a higher percentage of patients
abiraterone-prednisone over patients treated with predni­ with PSA responses (defined as reductions of at least 50%
sone monotherapy (not reached vs. 27.2 months) [12]. from baseline) in the lutetium-treated group [29]. This sug­
Enzalutamide’s efficacy in mCRPC was shown as it improved gested that 177Lu-PSMA-617 may have a role as an alternative
median OS compared to placebo in post-chemotherapy to cabazitaxel in mCRPC. Adverse events (AEs) of particular
mCRPC patients (18.4 months vs. 13.6 months) [13] and interest in this study include dry mouth, dry eyes, diarrhea,
then in the pre-chemotherapy mCRPC setting where there thrombocytopenia, and neuropathy. Dry mouth and dry eyes
was a radiographic progression-free survival (rPFS) and OS were seen in 60% and 30% of the patients treated with
benefit in the enzalutamide group compared to pla­ lutetium as compared to 21% and 4%, respectively, of patients
cebo [14]. treated with cabazitaxel. This was all grade 1–2 with no grade
In non-metastatic CRPC (nmCRPC), apalutamide, enzalu­ 3–4 events seen for either treatment with regard to these AEs.
tamide, and darolutamide have all been shown to improve Diarrhea was seen in approximately triple the percentage of
metastasis-free survival as the primary endpoint. The patients receiving cabazitaxel as compared to lutetium, but
SPARTAN, PROSPER, and ARAMIS trials provided the sup­ again this was mostly grade 1–2. Thrombocytopenia was not
port for approval of apalutamide, enzalutamide, and daro­ only more frequent in lutetium but also more severe as 11% of
lutamide, respectively, in nmCRPC, specifically in patients patients treated with lutetium experienced this at grade 3–4
with PSA doubling times of ≤10 months [15–17]. All three compared to none of the patients treated with cabazitaxel.
registrational studies demonstrated significant improve­ Neuropathy, a known possible AE of taxane chemotherapy,
ments in OS as secondary endpoints. occurred in 26% of patients treated with cabazitaxel as com­
ARPIs have also been widely adopted in mCSPC. pared to lutetium, which is important in a population that may
Abiraterone with prednisone added to ADT improved OS in have residual neuropathy from prior taxane treatment. The
two separate randomized studies: LATITUDE and an arm of the phase III VISION trial enrolled patients with mCRPC after failure
STAMPEDE trial [18,19]. In LATITUDE, the median OS was not from at least one prior ARPI and one taxane treatment and
reached for the abiraterone group compared to 14.8 months compared 177Lu-PSMA-617 to SOC therapy. The improvement
in the placebo group. The STAMPEDE study showed fewer in OS in the 177Lu-PSMA-617 arm (median OS 15.3 vs. 11.3
deaths in the ADT plus abiraterone group compared to ADT months, HR 0.62, 95% CI, 0.52–0.74) led to U.S. FDA approval
alone: 184 versus 262 (HR 0.63; 95% CI, 0.52–0.76, P < 0.001). in March 2022 in this setting [30].
The ENZAMET trial showed OS improvement in the enzaluta­ Immunotherapy in prostate cancer has a limited role in the
mide plus standard of care (SOC) arm compared to the SOC setting of mCRPC. The only vaccine therapy in prostate cancer
arm (5-year OS 67% vs. 57%, HR 0.7; 95% CI, 0.58–0.84), with to demonstrate an OS benefit thus far is sipuleucel-T, with
docetaxel allowed as SOC [20,21]. Apalutamide with ADT also a median OS of 25.8 versus 21.7 months (HR 0.78, 95% CI,
yielded an OS advantage compared to ADT alone, with med­ 0.61–0.98), although there was no improvement in PFS or
ian OS not reached versus 52.2 months (HR 0.65, p < 0.0001) in PSA response [31]. This treatment is reserved for asympto­
the TITAN trial [22,23]. In the last year, the needle has moved matic or minimally symptomatic mCRPC patients without visc­
further for mCSPC treatment, with data supporting the use of eral metastases, and the magnitude of OS benefit was found
ARPIs in combination with docetaxel and ADT, termed triplet to be greater in patients with lower baseline PSA [32].
therapy. In the ARASENS trial, the addition of darolutamide Pembrolizumab, a PD-1 inhibitor, currently carries approvals
compared to placebo to docetaxel and ADT improved OS by only for prostate cancer patients with MSI-H and TMB-H dis­
an HR of 0.68; 95% CI, 0.57–0.80 [24]. Similarly, the addition of ease [33,34]. These approvals were based on tissue-agnostic
abiraterone with prednisone (vs. placebo) to docetaxel and solid tumor trials that did not have particularly large numbers
EXPERT REVIEW OF ANTICANCER THERAPY 627

of prostate cancer patients. Trials of immune checkpoint inhi­ treatment [42]. This trial randomized patients to either abira­
bitors in unselected prostate cancer patients have failed to terone and olaparib or abiraterone and placebo. The primary
demonstrate improvements in survival [35–37]. There is cur­ endpoint of imaging-based progression-free survival (ibPFS)
rently no indication beyond this biomarker-based population favored the combination group over placebo (24.8 vs. 16.6
especially given failure of phase III trials that included doce­ months, HR 0.66; 95% CI, 0.54–0.81, p < 0.001). A second study
taxel in the mCRPC space (Keynote-921: NCT03834506) and looking at the combination of PARPi with oral anti-androgens
with enzalutamide (Keynote 641: NCT03834493) as well as in mCRPC is TALAPRO-2. This phase III study randomized
metastatic hormone-sensitive prostate cancer setting patients to enzalutamide plus either talazoparib or placebo
(Keynote-991: NCT04191096). in first-line mCRPC [43]. The primary endpoint was the same as
Therapies targeting the DNA damage repair (DDR) path­ in PROpel: ibPFS. The median ibPFS again favored the oral
ways with poly-ADP ribose (PARP) inhibitors have also demon­ anti-androgen and PARPi combination arm over the anti-
strated efficacy in selected patients with mCRPC. Olaparib and androgen and placebo arm (NR vs 21.9 months, HR 0.63; 95%
rucaparib were examined in patients with mCRPC who pro­ CI, 0.51–0.78; p < 0.001). Importantly, neither study allowed for
gressed on prior ARPIs in the phase III, PROfound and phase II, patients on the placebo arm to receive PARPi at time of
TRITON studies, respectively [38–40]. Olaparib was studied in progression, a comparison that would mirror a current
the phase III PROfound trial with the primary endpoint of approach available to selected patients in the United States
investigator-assessed imaging PFS in those who had at least who progress on abiraterone in the mCRPC setting.
one homologous recombination repair (HRR) mutations that There are many ongoing trials involving 177Lu-PSMA-617
included BRCA1, BRCA2, or ATM and failed prior ARPI in cohort that may expand its role further beyond mCRPC. PSMAfore
A. Other DDR mutations were included in cohort B. (NCT04689828) is a phase III trial in taxane-naive mCRPC
Conversely, the TRITON study required progression on one patients progressing on one line of ARPIs, and who are eligible
prior taxane and limited HRR mutations to BRCA1 and BRCA2. for a change to a different ARPI [44]. Patients in PSMAfore will
Analysis from PROfound demonstrated a PFS and OS benefit in be randomized to either treatment with 177Lu-PSMA-617 or
favor of olaparib over ARPI in the BRCA1, BRCA2, and ATM an ARPI (abiraterone/enzalutamide) with the primary endpoint
cohort (median OS 19.1 vs. 14.7 months, HR 0.69; 95% CI, for the trial being rPFS. Another trial, PSMAddition
0.50–0.97), while that from TRITON showed antitumor activity (NCT04720157), is evaluating 177Lu-PSMA-617 in mCSPC
in the form of objective responses and PSA response in [45]. This trial will randomize patients to either 177Lu-PSMA
patients treated with rucaparib. Another PARP inhibitor stu­ -617 added to SOC (defined as ADT+ARPI) versus SOC alone
died in mCRPC was niraparib in the phase II GALAHAD trial, with a primary endpoint of rPFS. A phase 1b trial (PRINCE)
which included 289 patients of whom overall response rates in investigating the benefits of immunogenic priming with
the BRCA cohort (n = 79) was 34.2% [41]. Collectively, these 177Lu-PSMA-617 followed by pembrolizumab in chemother­
aforementioned trials led to the eventual U.S. FDA approval of apy-naive mCRPC patients [46] demonstrated a promising
olaparib in adult patients with deleterious or suspected dele­ overall response rate of 44% in the 18 patients enrolled and
terious germline or somatic HRR gene-mutated mCRPC, who has led to further phase II trial testing (NCT03805594). PSMA-
have progressed following prior treatment with enzalutamide based ligand forms are also being combined with lutetium in
or abiraterone on May 19, 2020. While rucaparib was granted phase III clinical trials for patients with mCRPC who have failed
U.S. FDA accelerated approval on May 15, 2020 and niraparib prior ARPI but remain chemotherapy naive. The two studies
was given a U.S. FDA breakthrough therapy designation exploring this includes the following studies: SPLASH
on October 4 2019, the approval was reserved only for (NCT04647526) and ECLIPSE (NCT05204927).
mCRPC patients with deleterious BRCA mutation (germline Alpha particles are attractive as these deliver high energy
and/or somatic) previously treated with ARPI and a taxane- with fewer particle tracks to effect cell kill, have a short depth
based chemotherapy. of penetration, and have shorter half-lives, potentially limiting
toxicity to normal tissues. Additionally, from a practical per­
spective, the easier production of alpha particles lends for
3. Emerging treatment options
wider dissemination and use in the clinic [47]. The only
There are several treatment candidates with various mechanisms alpha emitter approved thus far in prostate cancer is radium
of action being evaluated in prostate cancer. Most involve utiliz­ 223 dichloride. One alpha emitter being evaluated is actinium-
ing prostate-associated antigens and combining these with 225 (225Ac). A phase I study combining this with a PSMA-
other forms of treatment. The therapies discussed herein include localizing antibody, J591, was recently presented by Tagawa
combination PARPi and oral anti-androgens, radioligands, mono­ et al. (NCT03276572) [48,49]. Thirty-two patients with mCRPC
clonal antibodies, small molecules, cell membrane receptors, who progressed on at least 1 ARPI and chemotherapy were
chimeric antigen receptor T-cell (CAR-T), cytokine-based thera­ enrolled in the study. Interestingly, prior treatment with
pies, and antibody-drug conjugates (ADCs). radium 223 and lutetium was allowed, and PSMA PET positiv­
As noted above, PARPi are approved for treatment in select ity was not required for enrollment. There was only one
mCRPC patients. More recent trials have begun to examine its patient with dose-limiting toxicity (DLT) at all the planned
role in combination with oral anti-androgens in unselected dose levels: grade 4 anemia and thrombocytopenia. Grade 3
patients. The PROpel study is a phase III study combining or higher AEs were hematologic. Lower grade AEs included
abiraterone and olaparib in unselected mCRPC as a first-line fatigue, pain flare, nausea, and xerostomia (which occurred in
628 M. ATIQ ET AL.

8 patients – 5 previously treated with lutetium – and was responses occurred in 6 out of 10 patients in dose levels 5 and
limited to grade 1). Clinical data presented included 22 out 6 and 1 out of 18 patients had a RECIST response (at dose
of 32 patients having any decline in PSA and 12 out of 32 level 4). CRS was the most common AE, occurring in 27 out of
patients experiencing ≥50% PSA reduction (PSA50 response). 32 (84.3%) patients, mostly in the first 2 cycles, presenting
CAR-T cell therapy is a new area of therapy being evaluated with fever, transaminitis, and hypotension. DLTs included
in prostate cancer. A first-in-human phase I study of PSMA rash and GI hemorrhage, both reversible [54]. An example of
CAR-T by Narayan et al. (NCT03089203) enrolled mCRPC another BiTE (with its tumor antigen) engaging CD3 on T-cells
patients and treated them with a PSMA-directed armored in mCRPC clinical trials is AMG 509 (STEAP1)
CAR-T cell that was modified with a dominant-negative trans­ (NCT04221542) [55].
forming growth factor (TGF)-beta receptor [50]. This study Another mode of immunotherapy holding promise is cyto­
utilized a 3+3 safety design with and without lymphodeple­ kine-based therapies, which may achieve a broader immuno­
tion. Patients were dosed in four cohorts at a range of 10–­ logic impact beyond T-cells by having a pleiotropic effect on
30 million cells or 100–300 million cells with or without the tumor microenvironment, including NK cells, myeloid-
lymphodepletion. Of the 13 patients treated, 4 had PSA reduc­ derived suppressor cells, and tumor-associated macrophages
tions of at least 30%. It must be noted, however, that one of [56]. Clinical application of free cytokines has been largely
the four patients died in the context of grade 4 cytokine limited by significant toxicity, which often prevented delivery
release syndrome (CRS) and sepsis, emphasizing the need for of effective doses to the tumor [57–60]; however, develop­
continued vigilance regarding toxicity monitoring. A second ment of modified cytokines is hoped to improve tumor deliv­
phase I study by Dorff et al. (NCT03873805) looked at CAR-T ery and reduce toxicity. The multi-arm QuEST1 trial is
cell therapy targeted to prostate stem cell antigen (PSCA) in examining successive cumulative combinations of BNVax (a
mCRPC after at least abiraterone or enzalutamide [51]. Patients poxviral-based vaccine against brachyury) and bintrafusp
were also required to have disease that expressed PSCA as alpha (a novel PD-L1 inhibitor with a TGF-β trap) with N-803
confirmed by City of Hope’s Pathology Department. These (an IL-15 superagonist) and epacadostat (an indoleamine 2,3
CAR-T cells are also using the 4-1BB costimulatory molecule. (IDO1) inhibitor). Preliminary results show four out of nine
Four cohorts were planned for treatment with cohort 1 receiv­ (44%) patients in the BNVax plus bintrafusp alpha and N-803
ing 100 million cells without lymphodepletion chemotherapy. arm having a PSA reduction by at least 30%, and two out of
The other three cohorts were planned to receive lymphode­ three (66%) evaluable patients having a radiological response
pletion first followed by 100 million, 300 million, or 600 million (compared to 1/13 (7.8%) and 0/3, respectively, in the BNVax
cells per each cohort, respectively. This first-in-human study and bintrafusp alpha only arm), possibly indicating activity of
encountered two DLTs (grade 3 noninfective cystitis and fati­ N-803 [61]. NHS-IL12 is an immunocytokine composed of two
gue in both) at a dose of 100 million cells after lymphodeple­ IL-12 heterodimers fused to an NHS76 antibody that targets
tion chemotherapy. However, upon amendment of the exposed DNA in areas of necrosis. A phase I/II trial combining
lymphodepletion chemotherapy to reduce the cyclophospha­ NHS-IL12 with docetaxel in mCSPC and mCRPC is ongoing
mide portion, no DLTs were encountered in an additional (NCT04633252) [62]. Preliminary data have established the
three patients. All three patients treated at the 100 million safety of the combination and the RP2D.
cell dose level without lymphodepletion progressed, while ADCs are another area being explored in prostate cancer.
seven of the nine patients treated at the same cell number These ADCs include antigen targets, such as six-
but with lymphodepletion had stable disease. The data transmembrane epithelial antigen of prostate 1 (STEAP-1),
acquired at this level allowed for escalation to the next dose trophoblast antigen 2 (TROP2), PSMA, and B7 homolog 3 (B7-
level of 300 million cells. H3). A phase I trial of an ADC (DSTP3086S) comprising
Another form of antibody-focused treatment in develop­ a humanized IgG1 STEAP-1 monoclonal antibody linked to
ment utilizes Bi-specific T-cell engagers (BiTEs). An example is monomethyl-auristatin E was conducted in mCRPC [63]. The
pasotuxizumab, which engages CD3 on T-cells and PSMA on 3+3 dose escalation study treated 77 patients. DLTs included
prostate cancer cells. A phase I trial enrolled 47 patients with transaminitis, hyperglycemia, and hypophosphatemia. Of the
mCRPC post ≥1 taxane and either abiraterone or enzaluta­ 62 patients who were treated at doses >2 mg/kg, 11 (18%) had
mide, and escalated doses of pasotuxizumab in subcutaneous a PSA50 response. This evidence of antitumor activity compli­
(SC) and continuous intravenous infusion (cIV) routes [52]. It mented the tolerable safety profile in this clinical trial
demonstrated activity of the treatment, with dose-dependent (NCT01283373). Sacituzumab govitecan (a humanized anti­
PSA reductions. Overall, PSA50 responses occurred in 9 out of body to TROP2 combined with SN-38, the active portion of
31 (29%) patients in the SC arm and 3 out of 16 (18.8%) in the irinotecan) is being evaluated in a phase II clinical trial of
cIV arm. Common AEs in the SC and cIV arms, respectively, mCRPC patients with progression on abiraterone or enzaluta­
included fever (25/31 [81%] and 15/16 [94%]), injection-site mide (NCT03725761) [64]. This ongoing study will enroll
reactions (24/31 [77%] and 0/16 [0%]), chills (7/31 [23%] and patients at a dose of 10 mg/kg and the primary endpoint will
11/16 [69%]), and fatigue (11/31 [36%] and 5/16 [31%]). be rPFS. PSMA-focused ADCs include one form, conjugated
Despite promising activity, 30 out of 30 evaluable patients with monomethyl-auristatin E, which has been tested in the
developed anti-drug antibodies, prompting development of mCRPC setting in a phase II study (NCT01695044) [65]. In this
an improved compound, AMG160 [52,53]. In turn, a phase trial, 119 mCRPC patients who had progressed on abiraterone
I trial of AMG160 monotherapy in mCRPC post ≥1 taxane or enzalutamide were treated with the PSMA ADC. PSA50
and ARPI enrolled 32 patients treated at 6 dose levels. PSA50 responses were seen in 14% of all treated patients. Grade 3
EXPERT REVIEW OF ANTICANCER THERAPY 629

AEs that were the most common included fatigue, neuropa­ who are deemed to benefit. For instance, further subgroup
thy, anemia, neutropenia, and electrolyte imbalance. Another analysis of the ARASENS trial has shown no major difference
B7-H3 monoclonal antibody, DS-7300, which has a exatecan between OS outcomes in either high-volume or low-volume
derivative payload, has been explored in a phase I/II study of disease or in high-risk or low-risk disease, benefiting all
advanced solid tumors, including mCRPC (NCT04145622). The equally. However, as with any new treatment, there are
mCRPC subset included a heavily pretreated population of 29 challenges that will need to be dealt with. One ongoing
patients [66]. No patients had a treatment-emergent AE lead­ challenge is the sequencing of therapies. Treatments with
ing to discontinuation. Besides establishing an RP2D, prelimin­ overlapping toxicities or somewhat similar mechanisms
ary results from this study regarding the mCRPC cohort comprise many of the currently approved as well as the
demonstrated 6 patients with partial responses and 15 with developing options discussed above. Understanding what
stable disease. MGC018, which also targets B7-H3, but has appropriate sequence of treatments to maximize survival
a duocarmycin payload, was studied in a phase I multicohort benefits while optimizing the quality of life will be key to
expansion that included 26 out of 40 patients with prostate utilizing the treatments in a meaningful way. Additionally,
cancer [67]. At the last follow-up, 11 out of 22 patients with as treatments may share overarching mechanisms, cross-
mCRPC had ≥50% PSA reduction and 4 out of 7 patients had resistance may emerge that could reduce the utility of
radiographic tumor response and reduction from 13% to 35%. some of the available options. Sequencing of different treat­
ment options in the mCRPC space includes prioritization of
PARPi use over ARPIs or chemotherapy in those who harbor
4. Conclusion DNA repair gene (DDR) mutations especially BRCA1 and
BRCA2. However, there is also emerging benefit in combi­
Emerging treatments are moving beyond the mostly andro­
nation therapy with ARPI and PARPi irrespective of DDR
gen-focused regimens that have dominated the approved
mutations given synergistic responses and likely will
therapeutic strategies for years. There is a large focus on
become approved soon, though not yet as of this writing.
utilizing more therapies targeted to tumor antigens by explor­
Logistical challenges cannot be ignored as was already seen
ing rational combinations. As has been practiced in the past,
with the issues regarding the use of lutetium in 2022, with
most novel treatments are first evaluated in mCRPC and only
ongoing shortage. Utilization of such an agent on a broader
brought into earlier disease states if efficacy is proven; how­
scale would require increases in capacity for production;
ever, this need not be the case as long as the risk-to-benefit
a principle that should be considered with regard to many
ratio for use in earlier disease stages can be scientifically
of the emerging treatments mentioned above [68]. Finally,
justified. New therapeutics bring the promise of expanding
economic factors which have always been a significant chal­
options and improving survival for patients in the most
lenge in the implementation of new treatments will be
advanced stages of disease. There are many practical chal­
another variable that is important to consider. However,
lenges that can arise in the face of this, and creative strategies
despite advances in therapy for mCRPC, treatment
will need to be implemented to overcome them.
responses lack durability especially when progression
ensues. Therefore, a search for other novel therapies with
5. Expert opinion unique mechanism of action and better understanding of
mechanisms of resistance to newer treatment options such
With a multitude of potential options arising in the arma­ as lutetium must be sought.
ment of prostate cancer therapy, the future of treatment is
promising. In light of the current approach to treatment
focusing on the androgen receptor, alternative mechanisms Funding
of action looking beyond this are crucial to move the field This paper received no funding.
forward. Moreover, in light of a trend of intensification of
treatment as an approach to the mCSPC state, having more
treatment options may allow for greater latitude in dein­
Declaration of interest
tensification of existing treatment regimens, consequently
allowing for improved quality of life. On the other hand, JAC serves on Speakers’ Bureau of Astellas/Seagen, BMS, and Pfizer/
EMD Serono; Consulting/Advisory Board Role for Bayer, Janssen,
multiple factors need to be considered regarding the elig­
Astellas, Sanofi/Genzyme, Merck, Pfizer/EMD Serono, AVEO, and
ibility for treatment intensification for patients with mCSPC Immunomedics.
including volume of disease (high volume or low volume), The authors have no other relevant affiliations or financial involvement
risk of disease (high risk vs. low risk), and even use of with any organization or entity with a financial interest in or financial
stereotactic radiotherapy or primary radiotherapy in the conflict with the subject matter or materials discussed in the manuscript
apart from those disclosed.
prostate area in patients with oligometastatic disease. One
of the criteria for triplet therapy, beyond volume or high-
risk disease, includes whether patients are considered che­
motherapy-fit and likely remain to be the appropriate can­ Reviewer disclosures
didates for intensification therapy. Current trials continue to A reviewer on this paper is an occasional advisory board member for
further elucidate the appropriate population of patients Merck Sharp & Dohme, AstraZeneca, Ipsen, and Pfizer.
630 M. ATIQ ET AL.

ORCID 20. Davis ID, Martin AJ, Zielinski RR, et al. Updated overall survival
outcomes in ENZAMET (ANZUP 1304), an international, cooperative
Elias Chandran http://orcid.org/0000-0002-0150-7765 group trial of enzalutamide in metastatic hormone-sensitive pros­
Jeanny B. Aragon-Ching http://orcid.org/0000-0002-6714-141X tate cancer (mHSPC). J Clin Oncol. 2022;40(17_suppl):LBA5004–
LBA5004. doi:10.1200/JCO.2022.40.17_suppl.LBA5004.
21. Davis ID, Martin AJ, Stockler MR, et al. Enzalutamide with standard
References first-line therapy in metastatic prostate cancer. N Engl J Med.
2019;381(2):121–131. DOI:10.1056/NEJMoa1903835
1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA
22. Chi KN, Agarwal N, Bjartell A, et al. Apalutamide for metastatic,
Cancer J Clin. 2022 Jan;72(1):7–33.
castration-sensitive prostate cancer. N Engl J Med. 2019 Jul 4;381
2. Siegel DA, O’Neil ME, Richards TB, et al. Prostate cancer incidence
(1):13–24.
and survival, by stage and race/ethnicity - United States,
23. Chi KN, Chowdhury S, Bjartell A, et al. Final analysis results from
2001-2017. MMWR Morb Mortal Wkly Rep. 2020 Oct 16;69
TITAN: a phase III study of apalutamide (APA) versus placebo (PBO)
(41):1473–1480.
in patients (pts) with metastatic castration-sensitive prostate cancer
3. Han M, Partin AW, Zahurak M, et al. Biochemical (prostate spe­
(mCSPC) receiving androgen deprivation therapy (ADT). J Clin Oncol.
cific antigen) recurrence probability following radical prostatect­
2021;39(6_suppl):11–11. doi:10.1200/JCO.2021.39.6_suppl.11.
omy for clinically localized prostate cancer. J Urol. 2003 Feb;169
24. Smith MR, Hussain M, Saad F, et al. Darolutamide and survival in
(2):517–523.
metastatic, hormone-sensitive prostate cancer. N Engl J Med. 2022
4. Huggins C, Stevens RE Jr., Hodges CV. Studies on prostate cancer: iI.
Mar 24;386(12):1132–1142.
The effects of castration on advanced carcinoma of the prostate
gland. Arch Surg. 1941;43(2):209–223. 25. Fizazi K, Foulon S, Carles J, et al. Abiraterone plus prednisone
5. Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or added to androgen deprivation therapy and docetaxel in de
mitoxantrone plus prednisone for advanced prostate cancer. novo metastatic castration-sensitive prostate cancer (PEACE-1):
N Engl J Med. 2004 Oct 7;351(15):1502–1512. a multicentre, open-label, randomised, phase 3 study with a 2 × 2
6. Sweeney CJ, Chen YH, Carducci M, et al. Chemohormonal therapy factorial design. Lancet. 2022;399(10336):1695–1707. DOI:10.1016/
in metastatic hormone-sensitive prostate cancer. N Engl J Med. S0140-6736(22)00367-1
2015 Aug 20;373(8):737–746. 26. Parker C, Nilsson S, Heinrich D, et al. Alpha emitter radium-223 and
7. James ND, Sydes MR, Clarke NW, et al. Addition of docetaxel, survival in metastatic prostate cancer. N Engl J Med. 2013 Jul
zoledronic acid, or both to first-line long-term hormone therapy 18;369(3):213–223.
in prostate cancer (STAMPEDE): survival results from an adaptive, 27. Pandit-Taskar N, Batraki M, Divgi CR. Radiopharmaceutical therapy
multiarm, multistage, platform randomised controlled trial. Lancet. for palliation of bone pain from osseous metastases. J Nucl Med.
2016 Mar 19;387(10024):1163–1177. 2004 Aug;45(8):1358–1365.
8. de Bono JS, Oudard S, Ozguroglu M, et al. Prednisone plus cabazi­ 28. Seider MJ, Pugh SL, Langer C, et al. Randomized phase III trial to
taxel or mitoxantrone for metastatic castration-resistant prostate evaluate radiopharmaceuticals and zoledronic acid in the palliation
cancer progressing after docetaxel treatment: a randomised of osteoblastic metastases from lung, breast, and prostate cancer:
open-label trial. Lancet. 2010 Oct 2;376(9747):1147–1154. report of the NRG Oncology RTOG 0517 trial. Ann Nucl Med. 2018
9. De Wit R, De Bono J, Sternberg CN, et al. Cabazitaxel versus Oct;32(8):553–560.
abiraterone or enzalutamide in metastatic prostate cancer. N Engl 29. Hofman MS, Emmett L, Sandhu S, et al. [(177)lu]lu-PSMA-617 versus
J Med. 2019;381(26):2506–2518. DOI:10.1056/NEJMoa1911206 cabazitaxel in patients with metastatic castration-resistant prostate
10. de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and cancer (TheraP): a randomised, open-label, phase 2 trial. Lancet.
increased survival in metastatic prostate cancer. N Engl J Med. 2021 Feb 27;397(10276):797–804.
2011 May 26;364(21):1995–2005. 30. Sartor O, de Bono J, Chi KN, et al. Lutetium-177-PSMA-617 for
11. Fizazi K, Scher HI, Molina A, et al. Abiraterone acetate for treatment metastatic castration-resistant prostate cancer. N Engl J Med.
of metastatic castration-resistant prostate cancer: final overall sur­ 2021 Sep 16;385(12):1091–1103.
vival analysis of the COU-AA-301 randomised, double-blind, 31. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunother­
placebo-controlled phase 3 study. Lancet Oncol. 2012 Oct;13 apy for castration-resistant prostate cancer. N Engl J Med. 2010 Jul
(10):983–992. 29;363(5):411–422.
12. Ryan CJ, Smith MR, de Bono JS, et al. Abiraterone in metastatic 32. Schellhammer PF, Chodak G, Whitmore JB, et al. Lower baseline
prostate cancer without previous chemotherapy. N Engl J Med. prostate-specific antigen is associated with a greater overall survi­
2013 Jan 10;368(2):138–148. val benefit from sipuleucel-T in the immunotherapy for prostate
13. Scher HI, Fizazi K, Saad F, et al. Increased survival with enzaluta­ adenocarcinoma treatment (IMPACT) trial. Urology. 2013 Jun;81
mide in prostate cancer after chemotherapy. N Engl J Med. 2012 (6):1297–1302.
Sep 27;367(13):1187–1197. 33. Marcus L, Lemery SJ, Keegan P, et al. FDA approval summary:
14. Beer TM, Armstrong AJ, Rathkopf DE, et al. Enzalutamide in meta­ pembrolizumab for the treatment of microsatellite instability-high
static prostate cancer before chemotherapy. N Engl J Med. 2014 Jul solid tumors. Clin Cancer Res. 2019 Jul 1;25(13):3753–3758.
31;371(5):424–433. 34. Marabelle A, Fakih M, Lopez J, et al. Association of tumour muta­
15. Smith MR, Saad F, Chowdhury S, et al. Apalutamide treatment and tional burden with outcomes in patients with advanced solid
metastasis-free survival in prostate cancer. N Engl J Med. 2018 Apr tumours treated with pembrolizumab: prospective biomarker ana­
12;378(15):1408–1418. lysis of the multicohort, open-label, phase 2 KEYNOTE-158 study.
16. Fizazi K, Shore N, Tammela TL, et al. Darolutamide in nonmeta­ Lancet Oncol. 2020 Oct;21(10):1353–1365.
static, castration-resistant prostate cancer. N Engl J Med. 2019 Mar 35. Beer TM, Kwon ED, Drake CG, et al. Randomized, double-blind,
28;380(13):1235–1246. phase iii trial of ipilimumab versus placebo in asymptomatic or
17. Hussain M, Fizazi K, Saad F, et al. Enzalutamide in men with non­ minimally symptomatic patients with metastatic
metastatic, castration-resistant prostate cancer. N Engl J Med. 2018 chemotherapy-naive castration-resistant prostate cancer. J Clin
Jun 28;378(26):2465–2474. Oncol. 2017;35(1):40–47. DOI:10.1200/JCO.2016.69.1584
18. Fizazi K, Tran N, Fein L, et al. Abiraterone plus prednisone in 36. Kwon ED, Drake CG, Scher HI, et al. Ipilimumab versus placebo after
metastatic, castration-sensitive prostate cancer. N Engl J Med. radiotherapy in patients with metastatic castration-resistant pros­
2017 Jul 27;377(4):352–360. tate cancer that had progressed after docetaxel chemotherapy
19. James ND, de Bono JS, Spears MR, et al. Abiraterone for prostate (CA184-043): a multicentre, randomised, double-blind, phase 3
cancer not previously treated with hormone therapy. N Engl J Med. trial. Lancet Oncol. 2014;15(7):700–712. DOI:10.1016/S1470-
2017 Jul 27;377(4):338–351. 2045(14)70189-5
EXPERT REVIEW OF ANTICANCER THERAPY 631

37. Sweeney CJ, Gillessen S, Rathkopf D, et al. Abstract CT014: metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol.
iMbassador250: a phase III trial comparing atezolizumab with enza­ 2022;40(6_suppl):91–91. doi:10.1200/JCO.2022.40.6_suppl.091.
lutamide vs enzalutamide alone in patients with metastatic 52. Hummel HD, Kufer P, Grüllich C, et al. Pasotuxizumab, a BiTE( ) ®
castration-resistant prostate cancer (mCRPC). Cancer Res. 2020;80(16 immune therapy for castration-resistant prostate cancer: phase I,
Supplement):CT014–CT014. doi:10.1158/1538-7445.AM2020-CT014. dose-escalation study findings. Immunotherapy. 2021 Feb;13
38. de Bono J, Mateo J, Fizazi K, et al. Olaparib for metastatic (2):125–141.
castration-resistant prostate cancer. N Engl J Med. 2020 May 53. Kamat NV, Yu EY, Lee JK. BiTE-ing into prostate cancer with bispe­
28;382(22):2091–2102. cific T-cell engagers. Clin Cancer Res. 2021;27(10):2675–2677.
39. Abida W, Patnaik A, Campbell D, et al. Rucaparib in men with 54. Tran B, Horvath L, Dorff T, et al. 609O results from a phase I study of
metastatic castration-resistant prostate cancer harboring a BRCA1 AMG 160, a half-life extended (HLE), PSMA-targeted, bispecific
or BRCA2 gene alteration. J Clin Oncol. 2020 Nov 10;38 ®
T-cell engager (BiTE ) immune therapy for metastatic castration-
(32):3763–3772. resistant prostate cancer (mCRPC). Ann Oncol. 2020;31:S507.
40. Hussain M, Mateo J, Fizazi K, et al. Survival with Olaparib in meta­ 55. Danila DC, Waterhouse DM, Appleman LJ, et al. A phase 1 study of
static castration-resistant prostate cancer. N Engl J Med. 2020;383 AMG 509 in patients (pts) with metastatic castration-resistant pros­
(24):2345–2357. DOI:10.1056/NEJMoa2022485 tate cancer (mCRPC). J Clin Oncol. 2022;40(16_suppl):TPS5101–
41. Smith MR, Scher HI, Sandhu S, et al. Niraparib in patients with TPS5101. doi:10.1200/JCO.2022.40.16_suppl.TPS5101.
metastatic castration-resistant prostate cancer and DNA repair 56. Chandran E, Meininger L, Karzai F, et al. Signaling new therapeutic
gene defects (GALAHAD): a multicentre, open-label, phase 2 trial. opportunities: cytokines in prostate cancer. Expert Opin Biol Ther.
Lancet Oncol. 2022 Mar;23(3):362–373. 2022 Oct 03;22(10):1233–1243.
42. Clarke NW, Armstrong AJ, Thiery-Vuillemin A, et al. Abiraterone and 57. Ruef C, Coleman DL. Granulocyte-macrophage colony-stimulating
olaparib for metastatic castration-resistant prostate cancer. NEJM factor: pleiotropic cytokine with potential clinical usefulness. Rev
Evi. 2022;1(9):EVIDoa2200043. Infect Dis. 1990;12(1):41–62.
43. Agarwal N, Azad A, Carles J, et al. TALAPRO-2: phase 3 study of 58. Cohen J. IL-12 deaths: explanation and a puzzle. Science. 1995;270
talazoparib (TALA) + enzalutamide (ENZA) versus placebo (PBO) + (5238):908–908. DOI:10.1126/science.270.5238.908.a.
ENZA as first-line (1L) treatment in patients (pts) with metastatic 59. Siegel JP, Puri RK. Interleukin-2 toxicity. J Clin Oncol. 1991;9
castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2023;41 (4):694–704.
(6_suppl):LBA17–LBA17. doi:10.1200/JCO.2023.41.6_suppl.LBA17. 60. Maas RA, Dullens HFJ, Den Otter W. Interleukin-2 in cancer treat­
44. Sartor AO, Morris MJ, Chi KN, et al. Psmafore: a phase 3 study to ment: disappointing or (still) promising? A review. Cancer Immunol
compare 177Lu-PSMA-617 treatment with a change in androgen Immunother. 1993;36(3):141–148.
receptor pathway inhibitor in taxane-naïve patients with metastatic 61. Redman JM, Madan RA, Karzai F, et al. 616MO efficacy of BN-brachyury
castration-resistant prostate cancer. J Clin Oncol. 2022;40(6_suppl): (BNVax) + bintrafusp alfa (BA) + N-803 in castration-resistant prostate
TPS211–TPS211. doi:10.1200/JCO.2022.40.6_suppl.TPS211. cancer (CRPC): results from a preliminary analysis of the quick efficacy
seeking trial (QuEST1). Ann Oncol. 2020;31:S511.
45. Sartor AO, Tagawa ST, Saad F, et al. Psm addition: a phase 3 trial to
62. Atiq MO, Chandran EBA, Meininger L, et al. Safety evaluation of M9241
compare treatment with 177Lu-PSMA-617 plus standard of care
in combination with docetaxel in metastatic prostate cancer. J Clin
(SOC) versus SOC alone in patients with metastatic
Oncol. 2022;40(6_suppl):93–93. doi:10.1200/JCO.2022.40.6_suppl.093.
hormone-sensitive prostate cancer. J Clin Oncol. 2022;40(6_suppl):
63. Danila DC, Szmulewitz RZ, Vaishampayan U, et al. Phase I study of
TPS210–TPS210. doi:10.1200/JCO.2022.40.6_suppl.TPS210.
DSTP3086S, an antibody-drug conjugate targeting six-transmembrane
46. Aggarwal RR, Sam SL, Koshkin VS, et al. Immunogenic priming with
epithelial antigen of prostate 1, in metastatic castration-resistant prostate
177Lu-PSMA-617 plus pembrolizumab in metastatic castration
cancer. J Clin Oncol. 2019 Dec 20;37(36):3518–3527.
resistant prostate cancer (mCRPC): a phase 1b study. J Clin Oncol.
64. Lang JM, Kyriakopoulos C, Slovin SF, et al. Single-arm, phase II
2021;39(15_suppl):5053–5053. doi:10.1200/JCO.2021.39.15_suppl.
study to evaluate the safety and efficacy of sacituzumab govitecan
5053.
in patients with metastatic castration-resistant prostate cancer who
47. Targeted Alpha Therapy Working G, Parker C, Lewington V, have progressed on second generation AR-directed therapy. J Clin
Shore N, et al. Targeted alpha therapy, an emerging class of cancer Oncol. 2020;38(6_suppl):TPS251–TPS251. doi:10.1200/JCO.2020.38.
agents: a review. JAMA Oncol. 2018 Dec 1;4(12):1765–1772. doi: 10. 6_suppl.TPS251.
1001/jamaoncol.2018.4044. 65. Petrylak DP, Vogelzang NJ, Chatta K, et al. PSMA ADC monotherapy
48. Tagawa ST, Osborne J, Fernandez E, et al. Phase I dose-escalation in patients with progressive metastatic castration-resistant prostate
study of PSMA-targeted alpha emitter 225Ac-J591 in men with cancer following abiraterone and/or enzalutamide: efficacy and
metastatic castration-resistant prostate cancer (mCRPC). J Clin safety in open-label single-arm phase 2 study. Prostate. 2020;80
Oncol. 2020;38(15_suppl):5560–5560. doi:10.1200/JCO.2020.38.15_ (1):99–108. DOI:10.1002/pros.23922
suppl.5560. 66. Patel MR, Johnson ML, Falchook GS, et al. DS-7300 (B7-H3
49. Tagawa ST, Sun M, Sartor AO, et al. Phase I study of 225Ac-J591 for DXd-ADC) in patients (pts) with metastatic castration-resistant
men with metastatic castration-resistant prostate cancer (mCRPC). prostate cancer (mCRPC): a subgroup analysis of a phase 1/2 multi­
J Clin Oncol. 2021;39(15_suppl):5015–5015. doi:10.1200/JCO.2021. center study. J Clin Oncol. 2022;40(6_suppl):87–87. doi:10.1200/
39.15_suppl.5015. JCO.2022.40.6_suppl.087.
50. Narayan V, Barber-Rotenberg JS, Jung IY, et al. PSMA-targeting 67. Shenderov E, Mallesara GHG, Wysocki PJ, et al. 620P MGC018, an
TGFbeta-insensitive armored CAR T cells in metastatic anti-B7-H3 antibody-drug conjugate (ADC), in patients with
castration-resistant prostate cancer: a phase 1 trial. Nat Med. 2022 advanced solid tumors: preliminary results of phase I cohort
Apr;28(4):724–734. expansion. Ann Oncol. 2021;32:S657–659.
51. Dorff TB, Blanchard S, Martirosyan H, et al. Phase 1 study of 68. Chandran E, Figg WD, Madan R. Lutetium-177-PSMA-617: a vision
PSCA-targeted chimeric antigen receptor (CAR) T cell therapy for of the future. Cancer Biol Ther. 2022 Dec 31;23(1):186–190.

You might also like