Simple Summary
Metastatic pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine neoplasms characterized by excess catecholamine production and secretion. This review summarizes current systemic therapy options and emerging strategies. Radiopharmaceuticals (131I-MIBG, 177Lu-DOTATATE) and cytotoxic regimens (CVD, temozolomide) remain cornerstone treatment options for disease control. Multi-targeted tyrosine kinase inhibitors (sunitinib, cabozantinib, axitinib) and the recently approved HIF-2α inhibitor belzutifan offer other targeted options, particularly for some PPGLs. Immune checkpoint inhibitors have modest activity, but combination approaches with TKIs or PARP inhibitors are currently under investigation. It is increasingly recognized that treatment for these tumors should be individualized based on tumor kinetics, genotype, and functional imaging phenotype, with multidisciplinary involvement and clinical trial participation being encouraged.
Abstract
Metastatic or unresectable pheochromocytomas and paragangliomas (PPGLs) remain rare but clinically challenging neuroendocrine neoplasms with limited curative options. Traditionally managed with surgery, radionuclide therapy, or cytotoxic chemotherapy, systemic treatments have historically achieved disease stabilization, rather than durable remissions. In recent years, however, the therapeutic landscape has evolved substantially. Radiopharmaceuticals such as 131I-MIBG and 177Lu-DOTATATE continue to play a pivotal role, achieving disease control in many patients. Cytotoxic regimens, particularly temozolomide, remain relevant, with some studies suggesting that SDHB-mutated PPGLs demonstrate a heightened sensitivity associated with MGMT promoter hypermethylation and reduced MGMT expression. Targeted agents are increasingly important: multi-kinase inhibitors such as sunitinib, anlotinib, and cabozantinib have shown meaningful activity. The landmark approval of belzutifan, a HIF-2α inhibitor, in 2025 represents the first oral targeted therapy for advanced/metastatic PPGL, which is particularly relevant for pseudohypoxic Cluster 1 tumors. Immunotherapy has yielded modest responses with checkpoint inhibitor monotherapy, but ongoing studies of dual checkpoint blockade and TKI–ICI combinations hold promise. Novel approaches, including PARP inhibition, metabolic targeting strategies, and cancer vaccines, are under investigation, especially for aggressive SDHB-related disease. Optimal sequencing of these therapies is emerging as a central challenge, with treatment strategies increasingly tailored to molecular genotype, tumor behavior, and functional imaging phenotype. This review summarizes current evidence and highlights ongoing clinical trials, underscoring a paradigm shift toward precision medicine and rational combination strategies. Collectively, these advances bring cautious optimism that metastatic PPGLs may soon become a more manageable chronic disease with improved survival and quality of life.
1. Introduction
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine neoplasms, with an estimated incidence of approximately 0.6 per 100,000 individuals per year. Selected cohorts show survival differences by metastatic presentation and suggest that cytoreductive surgery may improve outcomes in carefully chosen patients, underscoring the need for effective systemic therapies [1].
PPGLs are characterized by the overproduction of catecholamines, and often present with symptoms that mimic around 30 other medical conditions [2]. Common clinical manifestations include a diverse range of symptoms/signs, such as palpitations, hyperhidrosis, headaches, and anxiety. The overlap between these presentations and those of more prevalent disorders complicates the diagnostic process for PPGLs [3]. Clinicians typically evaluate three key considerations: (i) elevated levels of free plasma or urinary metanephrines (ii) evidence of the adrenal gland or retroperitoneal masses, and (iii) the presence of germline mutations in a PPGL susceptibility gene or specific molecular markers associated with the disease [4]. With approximately 20 known genetic mutations implicated in this condition, treatment approaches must be tailored to the individual patient [5]. Furthermore, the variability in patient presentations necessitates an understanding of how different individuals respond to the various levels of hormone secretion.
Surgical intervention is typically the first-line treatment for PPGL, and while there is some debate regarding the optimal surgical technique and timing, it is generally regarded as a curative option [6]. However, certain metastatic PPGLs may be unresectable, necessitating alternative therapeutic strategies. The timeline for implementing these alternatives is contingent upon disease progression and tumor burden, as well as the presence of mass effect. Patients exhibiting slow progression and low tumor burden are often placed under surveillance with no immediate therapeutic intervention, unless the disease involves a critical area with limited anatomical space, wherein earlier treatment may be warranted. Conversely, those with slow-to-moderate progression but moderate-to-high tumor burden are generally treated using radionuclide therapies such as peptide receptor radionuclide therapy (PRRT) and I131-Metaiodobenzylguanidine (131I-MIBG) therapy. In cases of rapid disease progression and significant visceral tumor burden, chemotherapy regimens may be instituted. Should these treatment strategies prove ineffective, patients may then be considered for therapies such as tyrosine kinase inhibitors [7]. If these options also yield insufficient results, participating in clinical trials may be recommended as a viable means of exploring additional treatment avenues that could potentially be beneficial for the disease [8].
This review aims to evaluate various treatment modalities and recent advances in systemic therapies for metastatic PPGLs. We integrate post-approval evidence for the HIF-2α inhibitor belzutifan in advanced PPGLs and discuss practical positioning relative to TKIs, radiopharmaceuticals, and chemotherapy. We synthesize the growing prospective and real-world PRRT evidence that is specific to PPGLs. Finally, we include a genotype–phenotype–therapy correlation table to facilitate personalized care
2. Definition of Pheochromocytoma and Paraganglioma (PPGL)
Pheochromocytoma and paraganglioma (PPGL) are rare neuroendocrine tumors that arise from chromaffin cells of the adrenal medulla or from the neural crest progenitors located outside of the adrenal gland [9]. According to the WHO Classification of Endocrine and Neuroendocrine Tumors, paragangliomas are non-epithelial neuroendocrine neoplasms that generally produce catecholamines and secrete them into the bloodstream [10]. Approximately 80–85% of tumors are located in the adrenal medulla (pheochromocytomas), while 15–20% arise in extra-adrenal sites (paragangliomas) [2]. Tumors from the sympathetic chain (thorax, abdomen, pelvis) are usually catecholamine-producing, whereas those from parasympathetic tissues in the head and neck region are typically non-secreting, with only ~4% producing catecholamines [2].
Histologically, sustentacular cells (S-100/SOX10 positive) surround tumor nests and may provide supportive diagnostic information, especially in the distinction of primary from metastatic tumors [10]. The Ki-67 proliferation index can be used to assess proliferative activity, and if high, it is suggestive of a more aggressive tumor, often reflecting metastatic ones. The criterion for malignancy in PPGLs is the presence of metastases at sites where chromaffin tissue is normally absent: currently only in bones or lymph nodes [11].
3. Genetic Risk Factors
PPGLs are among the most heritable tumors: 30–40% harbor germline pathogenic variants overall, with pediatric cases showing ~70–80% germline prevalence [12]. The most common hereditary drivers include SDHx (SDHA/SDHB/SDHC/SDHD/SDHAF2), VHL, RET, NF1, TMEM127, and MAX, among others; SDHx accounts for ~20% of all PPGLs [13].
Most genetic mutations linked to PPGLs can be divided into three clusters (see Table 1): pseudohypoxia (Cluster I), kinase signaling (Cluster II), and Wnt-signaling (Cluster III) [14]. Cluster I encompasses VHL, EPAS1, and TCA-cycle genes (SDHx, FH, MDH2, SLC25A11, etc.) [15]. Genetic mutations related to Cluster II include RET, NF1, TMEM127, MAX, and HRAS. Cluster III includes UBTF-MAML3 fusions and CSFE1 mutation [16].
Metastatic propensity is gene-specific, rather than cluster-wide: SDHB/A, HIF2A, and MAML3 (and to some degree, FH) confer high metastatic risk, whereas RET/NF1/TMEM127 are generally low-risk; MAX carries an intermediate risk (~15–20%) [17]. In children, presentation more often includes sustained hypertension, reflecting persistent catecholamine excess. Although research is limited, especially for PPGLs that belong to Cluster 3, it is thought that all current therapies can be potentially beneficial; however, they are not highly successful or mainly curative in PPGL patients of all cluster mutations [18]. The efficacy of treatment depends on more than the specific gene mutations, due to the individuality of the disease in each patient. Below is a table outlining the different clusters of mutations, along with their cluster characteristics and common gene mutations.
4. Local Therapeutic Approaches
4.1. Surgery
When PPGLs are localized, they can be surgically resected [19]. While surgery may be a potentially curative treatment for localized tumors, in metastatic disease, surgery is primarily palliative [7]. Despite this, a retrospective study with 113 patients showed an increase in the median overall survival from 36 to 148 months in patients with a metastatic disease undergoing debulking surgery [20]. Even if a tumor is localized, the size and location of the tumor can provide major difficulties in being able to effectively and safely resect the tumor. For adrenal tumors, unilateral adrenalectomy usually preserves adequate adrenal function; thus, lifelong steroid replacement is required only after bilateral adrenalectomy [21]. In hereditary/bilateral contexts, a partial adrenalectomy may reduce lifelong steroid dependence [22]. Despite these challenges, surgery is still one of the most effective ways to remove these tumors [23].
In preparation for surgery, alpha adrenoceptor blockers are initiated 7–14 days before for vasodilation and to decrease blood pressure, which can help regulate excessive hormone secretion [3]. Following alpha-blockade, beta blockers may be needed to help stabilize the patient’s cardiovascular system during surgery [4]. Calcium channel blockers are suitable adjuncts/alternatives in selected patients. Metyrosine, that is inhibiting catecholamine synthesis (50–80% reduction) [24], may be considered in patients with high catecholamine burden or suboptimal control.
4.2. Liver-Directed Therapies
Liver-directed therapies may be indicated in a subset of patients, particularly those with oligometastatic disease or a symptomatic lesion, with the intent to provide local control and palliation [25]. Liver tumors that are less than 2–3 cm can be more difficult to resect and those may be ablated [7]. Ablation is performed under ultrasound or CT guidance; device power and time are tailored (typical ranges ~60–150 W for ~3–10 min) [26,27,28].
A single center retrospective series reported different types of ablation of 123 metastases in 31 patients (24 PCC, 7 PPGL). These metastases were in various locations, including bone (51%), liver (44%), and other locations (5%). In this study, radiofrequency ablation (61%), cryoablation (33%) and percutaneous ethanol injection (6%) were used as treatments. The technical success was around 94%. A total of 80 of the metastases had imaging follow-ups, and among these, 86% achieved local control, which was equal for RFA and cryoablation. Local control was 94% in the liver (all RFAs), 88% bone cryoablation, and 74% for bone RFA. Pain control was achieved in 100% of the subjects [29].
An alternative to ablation is liver embolization. Transcatheter arterial embolization (TAE) is a type of liver embolization that is potentially used for decreasing free catecholamines. A few case studies have reported that TAE could perform control of hyper catecholamine secretion [30]. This procedure has been demonstrated as an acceptable bridge to elective surgical tumor resection [30]. Case reports describe rapid BP stabilization in catecholamine crisis after TAE, and in ruptured pheochromocytoma, tumor shrinkage enabled delayed elective surgery (e.g., reduction from 5.7 cm to ~1.0 cm at 6 months) [31]. While these data are promising, it is important to consider that further studies are needed to confirm the efficacy of TAE in reducing tumor size in ruptured pheochromocytomas.
5. Current Systemic Therapies for PPGL
5.1. Somatostatin Analogs
Neuroendocrine tumors tend to overexpress somatostatin receptors (SSTRs). In PPGL, SSTR expression is common at about 90% in metastatic disease, but varies by subtype [32]. In a cohort of 202 PPGLs patients, 50% were SSTR2 positive [33]. Another study observing 151 primary tumors found that over half of the tumors strongly expressed either SSTR2 or SSTR3 [34]. Patients with PPGLs can be treated with somatostatin analogs (SSAs), which can suppress hormone release and tumor cell proliferation [35], but high-level efficacy data with “cold” SSAs in PPGLs remain limited [32]. In practice, there are three main Somatostatin analogs (SSAs) used in treating PPGLs: octreotide, lanreotide, and pasireotide [36]. Antitumor efficacy and approval is established in GEP-NETs for Octreotide and Lanreotide (PROMID and Clarinet trial) [37,38], but there is limited PPGL- specific data. A dedicated Phase II in PPGLs (LAMPARA) is ongoing (see Table 2) [39].
Table 2.
Somatostatin analogs.
Pasireotide is the newest SSA and it targets somatostatin receptors SSTR1, SSTR2, SSTR3, and SSTR5 [40]. Additionally, Pasireotide has a 40-fold higher affinity for SSTR5, 30-fold higher for SSTR1, and 5-fold higher for SSTR3, compared to octreotide [41].
Pasireotide is approved for targeting acromegaly and Cushing’s, not NET/PPGL; in a Phase III study of refractory carcinoid syndrome, pasireotide did not outperform octreotide for symptom control, with exploratory signals for longer PFS [40].
5.2. Peptide Receptor Radionuclide Therapy PRRT
Peptide receptor radionuclide therapy (PRRT), with 177Lu-DOTATATE, is used to treat somatostatin-receptor-positive PPGLs and has shown activity with acceptable toxicity in multiple cohorts [42,43]. A nationwide retrospective study in Denmark, which included patients who had been diagnosed with PPGLs and had received PRRT over a 15 year period, showed that patients with a germline mutation, including SDHB, who received PRRT had better overall survival than patients without germline mutations (p = 0.041) [44]. The median follow-up time for this study was 31 months, and it showed an overall survival benefit for those with germline mutations. Patients with SDHB had a PFS of about 18 months, which was not achieved by patients with other mutations like SDHD or RET. Importantly, it should be noted that 177Lu-DOTATATE is associated with transient significant endocrine fluctuations and catecholamine surges, typically 24–48 h after administration, that are subsequently normalized. These potentially persistent effects are seen at a greater rate in more powerful agents, such as alpha particle PRRT [45]. A study performed in Stanford investigated six patients (one woman and five men) who expressed PPGL, four paraganglioma and two pheochromocytoma, using four 7.4 GBq cycles: PFS was 83%, 58%, and 39% at 11, 15, and 18 months, ORR was 40% at post-treatment assessment, disease control was 80% at 11 months, and mainly transient grade-3 cytopenias were without grade-3 hepatic/renal toxicity [46].
Prospectively, Lin et al. [47] conducted a phase-II Simon two-stage trial (four 7.4 GBq cycles; 36 evaluable) and reported a 6 month PFS rate of 86%, median PFS 20 months, and deeper responses over time (partial responses rising from 14% at end-of-treatment to 28% at follow-up). Outcomes differed by genotype: sporadic tumors had longer PFS (24.3 months) than SDHx tumors (12.9 months). Treatment was generally well tolerated but included 18% grade ≥ 3 catecholamine-release events.
In a case study with five patients, it was observed that when alpha/beta-blockers and calcium channel blockers were used to treat patients before their injections, there were no infusion reactions or catecholamine crises [48]. Overall, compared with “cold” SSAs (octreotide/lanreotide), PRRT in appropriately selected, SSTR-avid PPGLs provides higher rates of radiographic response and disease stabilization across a series, with an evolving but supportive safety profile [7,42].
5.3. Radioligand Therapy (MIBG)
PPGL frequently express the norepinephrine (NE) reuptake transporters on their cell surface. Meta-iodobenzylguinidine (MIBG) are substrates for these norepinephrine reuptake transporters that compete for binding sites and disrupt the NE reuptake mechanism; hence, reduction in the circulation of catecholamines. Highly specific MIBGs are labeled with radioactive iodine (131I-MIBG) and are used for specific targeted therapy [49]. In the pivotal phase II trial, patients received a dosimetric planning dose, followed by therapeutic doses of ~18.5 GBq (500 mCi) ~90 days apart. Among 64 evaluable patients, 92% achieved a partial response or stable disease within 12 months; 25% of all treated patients met the primary endpoint of a durable ≥50% reduction in antihypertensive medications. The median OS was ~36.7 months. Common AEs were myelosuppression, nausea, and fatigue, and no drug-related acute hypertensive crises occurred during/after infusion [50]. Overall, most patients treated with this drug achieved a decrease in tumor size, and lessened disease and symptoms as well. The findings showed a sustained control of catecholamine-associated hypertension in a quarter of the patients [51]. Not all PPGLs are sufficiently MIBG-avid for therapy, so avidity testing is essential [52]. Overall, for MIBG-avid disease, HSA 131I-MIBG achieves high rates of tumor control and biochemical/pressoric improvement in pretreated PPGL populations, often with a toxicity profile that is more manageable than multi-agent cytotoxic chemotherapy [53].
When both somatostatin SSTR and MIBG imaging demonstrate a therapeutically relevant uptake, the treatment selection should prioritize (1) dominant avidity and lesion-level heterogeneity, favoring the modality with stronger and more homogeneous uptake across sites; (2) bone marrow reserve and prior hematologic exposure, as both PRRT and 131I-MIBG can induce cytopenias; (3) renal function and the feasibility of amino acid nephroprotection with PRRT; (4) secretory status, given the transient catecholamine surges reported after PRRT that necessitate an optimal α/β blockade; (5) logistical and institutional radiation safety considerations; and (6) prior therapies and cumulative toxicity, to minimize additional marrow suppression when possible.
Head-to-head comparative data between PRRT (e.g., 177Lu-DOTATATE) and 131I-MIBG are lacking. No randomized trials directly compare these modalities, and the available evidence derives from small retrospective or nonrandomized series with inherent selection bias. In the largest PPGL cohort, PRRT was associated with longer overall and progression-free survival and higher response rates than 131I-MIBG, though results were confounded by referral and imaging bias [54]. A comparison reported a trend toward longer progression-free survival with 177Lu-DOTATATE versus 131I-MIBG (median of 29 vs. 18.5 months), but the difference was not statistically significant [55]. Other single-center series have similarly suggested improved outcomes with PRRT in select dual-avid PPGL cases, though these findings are limited by small sample sizes and nonuniform eligibility criteria [56]. Overall, direct comparative evidence remains absent, and existing data only suggest potential advantages for PRRT in subsets of dual-avid patients. Accordingly, therapy selection should remain individualized, and guided by imaging dominance, marrow reserve, renal and secretory status, prior therapies, and institutional expertise, while recognizing that PRRT and 131I-MIBG have distinct regulatory indications and toxicity profiles that complicate a direct comparison [57].
5.4. Tyrosine Kinase Inhibitors
Tyrosine kinases are critical mediators of oncogenic signaling pathways that regulate cancer cell proliferation, survival, and metastasis. Tyrosine kinase inhibitors (TKIs) represent a class of targeted agents that are designed to block aberrant kinase activity, thereby disrupting tumor growth and progression [58]. Patients with PPGLs have higher rates of angiogenesis and overexpression of growth factor receptors, such as vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFR) [8]. This is especially relevant in patients with Cluster 1 (pseudohypoxia) tumors, such as SDHx/VHL/EPAS1, where hypoxia signaling promotes angiogenesis. TKIs are thought to be effective in treating PPGLs in part due to their ability to inhibit angiogenesis [18].
TKIs are typically used to treat unresectable tumors and metastatic PPGLs. A meta-analysis demonstrated an ORR of 32%, a DCR 86%, and a median PFS of 8.9 months [59]. Common adverse effects of TKIs include hypertension, fatigue, diarrhea, and PPES (palmar-plantar erythrodysesthesia syndrome). While roughly one in seven patients do discontinue TKIs due to toxicity [59], it is common for patients to reduce their dosage to better tolerate adverse effects.
The most prevalent TKIs are sunitinib, axitinib, and cabozantinib (see Table 3) [60]. In progressive metastatic PPGLs specifically, the FIRSTMAPP randomized, double-blind phase II trial, met its primary endpoint with 12 month PFS 36% in the sunitinib group versus 19% in the placebo group [61,62]. The overall response rate to sunitinib for metastatic PPGLs is generally low; however, a small series suggests that SDHx-mutated tumors may respond to anti-angiogenic TKIs, and RET-altered pheochromocytomas have responded to selective RET inhibitors (e.g., selpercatinib), although robust predictive evidence for greater benefit with multi-target TKIs by genotype remains limited [63]. Retrospective data also note blood-pressure improvement in some patients on sunitinib [62].
On the other hand, a Phase 2 trial with axitinib (NCT01967576) indicated partial responses in three out of nine patients and stable disease in five out of nine [64]. For cabozantinib, the single-arm phase II NATALIE trial (NCT02302833) in progressive metastatic PPGLs showed an ORR of 25% and a median PFS of 16.6 months [65]. Responses were observed regardless of SDHB mutation status, indicating that cabozantinib is effective across both hereditary (SDHB-driven) and sporadic cases.
Additionally, anlotinib, a multi-target VEGFR/FGFR/PDGFR TKI (12 mg daily, 2 weeks on/1 week off), has shown encouraging real-world activity in advanced PPGLs, with ORR 44%, DCR 96%, and median PFS 25 months [66]; prospective phase II trials in PPGLs are ongoing (NCT04860700, NCT05133349).
Overall, while the ORR to single-agent TKIs is modest, clinically meaningful disease control is common, and dose modifications are frequently used to manage toxicity [59].
Table 3.
Tyrosine kinase inhibitors.
Table 3.
Tyrosine kinase inhibitors.
| Drug | Dose | Target | Patient Population Studied | Efficacy | Side Effects |
|---|---|---|---|---|---|
| Sunitinib [61] | 37.5 mg qd (FIRSTMAPPP), 50 mg qd also used. | VEGFR1–3, PDGFRα/β, KIT, FLT3, RET | Progressive/metastatic PPGLs (sporadic and germline) | FIRSTMAPPP (phase II RCT): 12 months PFS 36% vs. 19% (sunitinib vs. placebo). SUTNET (phase II, non-randomized): median PFS 14.1 months. | HTN, fatigue/asthenia, diarrhea. Grade ≥3 in FIRSTMAPPP: asthenia 18%, HTN 13%. |
| Cabozantinib [65] | 60 mg qd. | VEGFR2, MET, AXL, RET (multi-target) | Unresectable, progressive metastatic PPGL | NATALIE (phase II, single-arm): ORR 25%; median PFS 16.6 months; median OS 24.9 months. | HTN, hand-foot syndrome, dysgeusia, fatigue. |
| Axitinib [64] | 5 mg bid. | VEGFR1–3 (selective) | Progressive PPGL | Phase II (prelim.): PR 3/9, SD 5/9 (DCR ~89%). | HTN, fatigue; mucositis; Diarrhea. |
5.5. Chemotherapy (See Table 4)
5.5.1. Cyclophosphamide–Vincristine–Dacarbazine (CVD) Regimen
The most common forms of chemotherapy include cyclophosphamide, vincristine, and dacarbazine (CVD) and temozolomide (TMZ) to treat advanced or unresectable PPGL. In one of the earliest CVD reports, Keiser et al. described three patients with rapidly progressive malignant pheochromocytoma who experienced marked blood pressure reductions and clinical improvement within the first cycles of treatment [67].
Table 4.
Cytotoxic chemotherapy.
Table 4.
Cytotoxic chemotherapy.
| Drug/Regimen | Dose | Mechanism/Target | Study Group | ORR (in %), PFS (in Months), OS (in Months) | Side Effects |
|---|---|---|---|---|---|
| CVD (cyclophosphamide–vincristine–dacarbazine) | Cyclophosphamide 750 mg/m2 D1; vincristine 1.4 mg/m2 (max 2 mg) D1; dacarbazine 600 mg/m2 D1–2; q21–28 days (Averbuch protocol). | Cytotoxic combo: alkylator (cyclophosphamide), vinca (vincristine), alkylator (dacarbazine). | Metastatic/unresectable PPGL. | Small series show ORR 55–57%; meta-analysis (4 studies) CR 4%/PR 37%/SD 14%; 1 year PFS 45% in one cohort; OS inconsistently reported [68,69,70,71] | Mostly grade 1–2 myelosuppression, neuropathy, GI upset; rare hypertensive crises reported (optimize α/β-blockade). |
| Temozolomide (TMZ) | 150–200 mg/m2 PO daily D1–5 q28 days (monotherapy); in Kulke trial: TMZ 150 mg/m2 + thalidomide 100 mg daily. | Oral alkylating agent; activity linked to MGMT promoter hypermethylation (frequent in SDHB PPGL). | Metastatic PPGLs (retrospective series); mixed NET phase II included small PCC cohort. | Across PPGL series: PR 33–40%; median PFS 13.3 months–~26 months; 3 years OS 58% in one cohort. (NET phase II overall ORR 25%, median response 13.5 mo; PCC subset included) [72 73] | Generally well tolerated; lymphopenia/myelosuppression, fatigue, nausea. |
In a larger non-randomized, single arm trial by Averbuch et al. (n = 14), those diagnosed with metastatic PPGLs showed an overall response rate (ORR) of 57% and the trial showed complete or partial biochemical responses in 79% of patients, supporting activity of CVD in metastatic disease [68].
A 22 year follow-up study by Huang et al. (n = 18) reported CR 11% and PR 44% (ORR 55%), with consistent symptom and blood-pressure improvement and acceptable tolerability [69].
A systematic review and meta-analysis, pooling four studies (50 patients), estimated complete responses at 4%, partial responses at 37%, stable disease at 14% for tumor volume, and partial hormonal response at 40% (catecholamines) where reported, suggesting that CVD can palliate and shrink a tumor in a subset [70].
5.5.2. Temozolomide
Temozolomide (TMZ) is an oral alkylating chemotherapeutic again. A Phase II study was performed to determine the efficacy of TMZ in patients with metastatic NETs (n = 29). The radiologic response rate was 25% overall, with responses by subtype being 45% in pancreatic NETs, 33% in pheochromocytoma, and 7% in midgut NET; the median response duration was 13.5 months, 1 year OS was 79%, and 2 year OS was 61% [72]. Overall, the study showed that TMZ was an active treatment in pheochromocytoma. In metastatic PPGLs specifically, Hadoux et al. (n = 15) reported PR at 33%, all among SDHB-mutated tumors; median PFS at 13.3 months; and longer PFS for SDHB-mutated vs. non-mutated cases (19.7 vs. 2.9 months). SDHB status correlated with the MGMT promoter hypermethylation/low MGMT expression: a plausible mechanism for TMZ sensitivity [73].
A more recent retrospective series (Perez 2022, n = 19) found PR at 40% (6/15 evaluable), SD at 27%, PD at 33%, a median PFS of 2.2 years, and 3 year OS at 58%; in that cohort, outcomes did not favor SDHx carriers over noncarriers, highlighting heterogeneity across studies [74].
Pediatric data are limited, but a case report of a 12-year-old girl with SDHB-related metastatic PGL treated with TMZ after surgery documented clinical improvement and good tolerability, suggesting that TMZ can be considered in carefully selected children [75].
5.6. Immunotherapy in Advanced PPGL
In the first prospective trial of the PD-1 blockade, pembrolizumab monotherapy showed modest activity: only 1 of 11 patients (9%) achieved a partial response, but 40% of patients were progression-free at 27 weeks and the clinical benefit rate (including stable disease) reached about 72% [76]. Notably, treatment was tolerable with an acceptable safety profile. These results indicate that single-agent immunotherapy can stabilize disease in a subset of patients, though deep responses are rare. Dual checkpoint blockade is now under exploration; for example, a phase II trial combining nivolumab (anti–PD-1) with ipilimumab (anti–CTLA-4) in metastatic PPGLs is ongoing [77]. Early anecdotal reports also hint that combining immunotherapy with other agents may enhance efficacy. For instance, one case report described a dramatic and durable tumor regression when nivolumab was given together with the multi-kinase inhibitor cabozantinib, after multiple prior treatment failures [78].
Multiple datasets delineate a cluster-dependent immune contexture in PPGL. PD-L1 expression is generally low and tends to be lowest in pseudohypoxia-cluster tumors (SDHx, VHL, EPAS1), relative to kinase-signaling and sporadic cases, aligning with the modest activity seen in single-agent PD-1/PD-L1 blockade [79]. CD8+ T-cell infiltration shows a similar pattern (reduced in pseudohypoxic tumors) [80]; norepinephrine-secreting tumors also display lower PD-L1/CD8 than epinephrine-secreting diseases [81]. An exception is the MAML3-related subset, where PD-L1 positivity and a less immunosuppressive microenvironment have been reported [13]. This biology tempers expectations for a single-agent checkpoint blockade but does not preclude benefit in a subset of patients.
Future studies are focusing on combination approaches: pairing checkpoint inhibitors with targeted therapies or novel immunomodulators to improve response rates in this historically immunoresistant disease.
6. Novel Targeted Therapies: HIF-2α Inhibition and Other Strategies
A major advance in 2025 was the introduction of belzutifan, a selective inhibitor of hypoxia-inducible factor-2α (HIF-2α). In May 2025, the FDA approved belzutifan for adult and pediatric patients ≥12 y with locally advanced, unresectable, or metastatic PPGLs [82]. This approval was based on the LITESPARK-015 trial (n = 72 open label), in which belzutifan achieved a 26% objective response rate with a median duration of response (DOR) of 20.4 months; disease control was 85%, median PFS was 22.3 months, and 24 month OS was 76%. Among the 60 patients on baseline antihypertensives, 32% achieved a ≥50% sustained reduction in at least one medication for ≥6 months. Safety was manageable and consistent with prior experience; anemia was the most common grade ≥3 event (22%) [83,84]. Belzutifan targets the HIF-2α transcription factor, a known oncogenic driver in pseudohypoxic PPGLs [58]. This mechanism may be particularly relevant to SDHB-related tumors and other Cluster 1 PPGLs (e.g., VHL- and EPAS1-mutated), which are driven by dysregulated hypoxia signaling [58]. Indeed, HIF-2α inhibitors were long theorized as being promising agents for these patients, and belzutifan’s clinical success confirms that targeting the HIF pathway can yield meaningful tumor responses in advanced PPGL.
Going forward, belzutifan may provide a much-needed targeted option: it is an oral agent that can induce partial remissions and durable disease control, and it is being integrated into treatment algorithms, particularly for patients with Cluster 1 mutations.
Beyond HIF-2α, other molecular targets are under investigation. PARP inhibitors are one example: SDHB-mutant PPGLs accumulate oncometabolites (succinate) that may confer vulnerability to the PARP blockade [85]. Preclinical studies showed that adding the PARP inhibitor olaparib can amplify temozolomide’s cytotoxicity in SDHB-deficient models [58]. This has led to early-phase trials combining temozolomide with PARP inhibitors (e.g., talazoparib or veliparib) in metastatic PPGL. Another emerging strategy is exploiting the metabolic vulnerabilities of SDHB-related tumors. For instance, research is ongoing into targeting the unique metabolic pathways and redox state of SDHB-null cells (such as through oxidative stress induction or Krebs cycle enzyme modulation) [86]. While these approaches remain investigational, with belzutifan’s success, there is optimism that novel targeted agents, from HIF-2α antagonists to PARP inhibitors and beyond, can meaningfully improve outcomes.
7. Enhanced Treatment Sequencing and Future Directions
As of 2025, many active clinical trials are actively investigating treatments for PPGLs, many of which involve innovative combinations, such as cancer vaccines [87,88] with checkpoint inhibitors, novel immune drugs with PD-1 inhibitors, and multi-drug regimens incorporating PARP inhibitors, targeted agents, and chemotherapy [89,90]. The preliminary signals from these studies are encouraging, though full published data are awaited.
The expanding arsenal of systemic therapies has made treatment sequencing an increasingly nuanced aspect of PPGL care. An individualized approach is crucial, considering the tumor growth rate, disease burden, functional status, and tumor genotype or avidity (see Table 5).
Table 5.
Molecular clusters, imaging phenotype and therapeutic implications in PPGLs.
The optimal sequencing is not yet defined by guidelines, but multidisciplinary expert consensus emphasizes personalization [18]. Factors such as germline mutation status, tumor differentiation, and dominant imaging phenotype (MIBG-avid vs. SSTR-avid vs. FDG-avid) [91] guide therapy selection at each juncture.
Emerging biomarkers may further refine sequencing by indicating who is benefiting from a given treatment [25]. In all cases, the principle of maximizing disease control while minimizing cumulative toxicity prevails [51]. This often means using the least aggressive effective therapy first and escalating as needed, like starting with a radioligand or targeted agent before switching to multi-agent chemotherapy or experimental combinations. It also means enrolling patients in clinical trials whenever possible, so that they can access novel therapies.
As ongoing trials broaden our options, three real-world constraints warrant emphasis. First, therapeutic resistance: angiogenic escape with multi-target TKIs is common, and on-target resistance to HIF-2α inhibitors (e.g., EPAS1 G323E gatekeeper variants and the compensatory HIF-1 signaling described in RCC) may analogously limit durability in PPGLs [59,92]. Second, patient selection: lesion-level heterogeneity of SSTR/MIBG expression and the risk of catecholamine surges after PRRT require pre-treatment α/β-blockade, endocrine monitoring, and multidisciplinary planning [33,45]. Third, access and logistics: availability and reimbursement vary by region, and high-specific-activity 131I-MIBG production was discontinued in 2023 [93], which can influence sequencing choices. Priorities include trials integrating genotypes (e.g., SDHB, VHL/EPAS1), imaging phenotypes, and immune contextures, while capturing affordability and capacity constraints as new agents (e.g., belzutifan) move earlier in care.
8. Conclusions
Metastatic or unresectable pheochromocytomas and paragangliomas are uncommon but clinically challenging. The systemic armamentarium now spans radiopharmaceuticals (131I-MIBG and 177Lu-DOTATATE PRRT), cytotoxic chemotherapy (CVD and temozolomide), anti-angiogenic TKIs (e.g., sunitinib, cabozantinib, and axitinib), and the recently approved HIF-2α inhibitor belzutifan, with a single-agent immune checkpoint blockade showing modest activity. Optimal use of these modalities is individualized to tumor kinetics, germline/somatic genotype, and functional imaging phenotype (MIBG- or SSTR-avid disease), with attention to safety and local availability.
Looking ahead, priorities include biomarker-driven sequencing and rational combinations to overcome resistance and heterogeneity (e.g., PARP inhibitor–temozolomide strategies in SDHB disease, TKI–ICI combinations), as well as a pragmatic assessment of access, toxicity, and patient-reported outcomes. Prospective, genotype- and imaging-embedded trials remain essential to define comparative effectiveness and translate molecular insights into durable disease control and an improved quality of life for patients with PPGLs.
Author Contributions
Conceptualization: I.G., O.B., J.H.H.-F., and J.d.R.; resources I.G., O.B., J.H.H.-F., and J.d.R.; writing—original draft preparation, I.G., O.B., and J.H.H.-F.; writing, I.G., O.B., J.H.H.-F., K.P., F.I.L., and J.d.R.; supervision, K.P., F.I.L., and J.d.R. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
No new data were created or analyzed in this study. Data sharing is not applicable to this article.
Conflicts of Interest
The authors declare no conflicts of interest.
Abbreviations
The following abbreviations are used in this manuscript:
| 131I | Iodine-131 |
| 177Lu | Lutetium-177 |
| AE(s) | Adverse event(s) |
| AXL | AXL receptor tyrosine kinase |
| bid | Twice daily |
| CVD | Cyclophosphamide–Vincristine–Dacarbazine |
| D1, D1–5 | Day 1; Days 1 through 5 |
| DOTATATE | DOTA-Tyr3-octreotate (Lu-177 DOTATATE) |
| FLR/FLT3 | Fms-like tyrosine kinase 3 (FLT3) |
| GI | Gastrointestinal |
| GEP-NET | Gastroenteropancreatic neuroendocrine tumor |
| HFS | Hand-foot syndrome |
| HIF-2α | Hypoxia-inducible factor 2 alpha |
| HR | Hazard ratio |
| HTN | Hypertension |
| IM | Intramuscular |
| IRB | Institutional Review Board |
| KIT | KIT proto-oncogene receptor tyrosine kinase (CD117) |
| LAR | Long-acting release (e.g., octreotide LAR) |
| mg/m2 | Milligrams per square meter |
| MGMT | O6-methylguanine-DNA methyltransferase |
| MIBG | Meta-iodobenzylguanidine |
| mo | Months |
| MET | MET proto-oncogene receptor tyrosine kinase |
| NET | Neuroendocrine tumor |
| NIH | National Institutes of Health |
| ORR | Objective response rate |
| OS | Overall survival |
| PDGFR | Platelet-derived growth factor receptor |
| PFS | Progression-free survival |
| PO | By mouth (per os) |
| PPGL | Pheochromocytoma and Paraganglioma |
| PR | Partial response |
| PRRT | Peptide receptor radionuclide therapy |
| qd | Once daily |
| q21–28d/q28d | Every 21–28 days/every 28 days |
| RET | RET proto-oncogene receptor tyrosine kinase |
| SC | Subcutaneous |
| SD | Stable disease |
| SDHB | Succinate dehydrogenase complex iron sulfur subunit B |
| SI-NET | Small-intestinal neuroendocrine tumor |
| SSTR | Somatostatin receptor |
| SSA | Somatostatin analog |
| TKI | Tyrosine kinase inhibitor |
| TMZ | Temozolomide |
| TTP | Time to progression |
| VEGFR | Vascular endothelial growth factor receptor |
| α/β-blockade | Alpha/Beta-adrenergic blockade |
References
- Al Subhi, A.R.; Boyle, V.; Elston, M.S. Systematic Review: Incidence of Pheochromocytoma and Paraganglioma over 70 Years. J. Endocr. Soc. 2022, 6, bvac105. [Google Scholar] [CrossRef]
- Neumann, H.P.H.; Young, W.F.; Eng, C. Pheochromocytoma and Paraganglioma. N. Engl. J. Med. 2019, 381, 552–565. [Google Scholar] [CrossRef] [PubMed]
- Lenders, J.W.M.; Duh, Q.-Y.; Eisenhofer, G.; Gimenez-Roqueplo, A.-P.; Grebe, S.K.G.; Murad, M.H.; Naruse, M.; Pacak, K.; Young, W.F., Jr. Pheochromocytoma and Paraganglioma: An Endocrine Society Clinical Practice Guideline. J. Clin. Endocrinol. Metab. 2014, 99, 1915–1942. [Google Scholar] [CrossRef] [PubMed]
- Casey, R.T.; Hendriks, E.; Deal, C.; Waguespack, S.G.; Wiegering, V.; Redlich, A.; Akker, S.; Prasad, R.; Fassnacht, M.; Clifton-Bligh, R.; et al. International consensus statement on the diagnosis and management of phaeochromocytoma and paraganglioma in children and adolescents. Nat. Rev. Endocrinol. 2024, 20, 729–748. [Google Scholar] [CrossRef] [PubMed]
- Horton, C.; LaDuca, H.; Deckman, A.; Durda, K.; Jackson, M.; Richardson, M.E.; Tian, Y.; Yussuf, A.; Jasperson, K.; Else, T. Universal Germline Panel Testing for Individuals With Pheochromocytoma and Paraganglioma Produces High Diagnostic Yield. J. Clin. Endocrinol. Metab. 2022, 107, e1917–e1923. [Google Scholar] [CrossRef]
- Sukrithan, V.; Perez, K.; Pandit-Taskar, N.; Jimenez, C. Management of metastatic pheochromocytomas and paragangliomas: When and what. Curr. Probl. Cancer 2024, 51, 101116. [Google Scholar] [CrossRef]
- Fishbein, L.; Del Rivero, J.; Else, T.; Howe, J.R.; Asa, S.L.; Cohen, D.L.; Dahia, P.L.M.; Fraker, D.L.; Goodman, K.A.; Hope, T.A.; et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Management of Metastatic and/or Unresectable Pheochromocytoma and Paraganglioma. Pancreas 2021, 50, 469–493. [Google Scholar] [CrossRef]
- Wang, K.; Crona, J.; Beuschlein, F.; Grossman, A.B.; Pacak, K.; Nölting, S. Targeted Therapies in Pheochromocytoma and Paraganglioma. J. Clin. Endocrinol. Metab. 2022, 107, 2963–2972. [Google Scholar] [CrossRef]
- Pacak, K.; Wimalawansa, S.J. Pheochromocytoma and paraganglioma. Endocr. Pract. 2015, 21, 406–412. [Google Scholar] [CrossRef]
- Mete, O.; Asa, S.L.; Gill, A.J.; Kimura, N.; de Krijger, R.R.; Tischler, A. Overview of the 2022 WHO Classification of Paragangliomas and Pheochromocytomas. Endocr. Pathol. 2022, 33, 90–114. [Google Scholar] [CrossRef]
- Juhlin, C.C. Challenges in Paragangliomas and Pheochromocytomas: From Histology to Molecular Immunohistochemistry. Endocr. Pathol. 2021, 32, 228–244. [Google Scholar] [CrossRef]
- Amar, L.; Pacak, K.; Steichen, O.; Akker, S.A.; Aylwin, S.J.B.; Baudin, E.; Buffet, A.; Burnichon, N.; Clifton-Bligh, R.J.; Dahia, P.L.M.; et al. International consensus on initial screening and follow-up of asymptomatic SDHx mutation carriers. Nat. Rev. Endocrinol. 2021, 17, 435–444. [Google Scholar] [CrossRef]
- Calsina, B.; Piñeiro-Yáñez, E.; Martínez-Montes, Á.M.; Caleiras, E.; Fernández-Sanromán, Á.; Monteagudo, M.; Torres-Pérez, R.; Fustero-Torre, C.; Pulgarín-Alfaro, M.; Gil, E.; et al. Genomic and immune landscape Of metastatic pheochromocytoma and paraganglioma. Nat. Commun. 2023, 14, 1122. [Google Scholar] [CrossRef] [PubMed]
- Fishbein, L.; Leshchiner, I.; Walter, V.; Danilova, L.; Robertson, A.G.; Johnson, A.R.; Lichtenberg, T.M.; Murray, B.A.; Ghayee, H.K.; Else, T.; et al. Comprehensive molecular characterization of pheochromocytoma and paraganglioma. Cancer Cell 2017, 31, 181–193. [Google Scholar] [CrossRef] [PubMed]
- Calsina, B.; Currás-Freixes, M.; Buffet, A.; Pons, T.; Contreras, L.; Letón, R.; Comino-Méndez, I.; Remacha, L.; Calatayud, M.; Obispo, B.; et al. Role of MDH2 pathogenic variant in pheochromocytoma and paraganglioma patients. Genet. Med. 2018, 20, 1652–1662. [Google Scholar] [CrossRef]
- de Bresser, C.J.M.; de Krijger, R.R. The Molecular Classification of Pheochromocytomas and Paragangliomas: Discovering the Genomic and Immune Landscape of Metastatic Disease. Endocr. Pathol. 2024, 35, 279–292. [Google Scholar] [CrossRef]
- Kumar, S.; Lila, A.R.; Memon, S.S.; Sarathi, V.; Patil, V.A.; Menon, S.; Mittal, N.; Prakash, G.; Malhotra, G.; Shah, N.S.; et al. Metastatic cluster 2-related pheochromocytoma/paraganglioma: A single-center experience and systematic review. Endocr. Connect. 2021, 10, 1463–1476. [Google Scholar] [CrossRef]
- Nölting, S.; Bechmann, N.; Taieb, D.; Beuschlein, F.; Fassnacht, M.; Kroiss, M.; Eisenhofer, G.; Grossman, A.; Pacak, K. Personalized Management of Pheochromocytoma and Paraganglioma. Endocr. Rev. 2022, 43, 199–239. [Google Scholar] [CrossRef]
- Aygun, N.; Uludag, M. Pheochromocytoma and Paraganglioma: From Treatment to Follow-up. Şişli Etfal Hastan. Tip Bülteni 2020, 54, 391–398. [Google Scholar] [CrossRef]
- Roman-Gonzalez, A.; Zhou, S.; Ayala-Ramirez, M.; Shen, C.; Waguespack, S.G.; Habra, M.A.; Karam, J.A.; Perrier, N.; Wood, C.G.; Jimenez, C. Impact of Surgical Resection of the Primary Tumor on Overall Survival in Patients With Metastatic Pheochromocytoma or Sympathetic Paraganglioma. Ann. Surg. 2018, 268, 172–178. [Google Scholar] [CrossRef] [PubMed]
- Zawadzka, K.; Tylec, P.; Małczak, P.; Major, P.; Pędziwiatr, M.; Pisarska-Adamczyk, M. Total versus partial adrenalectomy in bilateral pheochromocytoma—A systematic review and meta-analysis. Front. Endocrinol 2023, 14, 1127676. [Google Scholar] [CrossRef]
- Pinto, D.; Parameswaran, R. Adrenal insufficiency post unilateral adrenalectomy in non-cortisol secreting tumours. Gland. Surg. 2023, 12, 1328–1331. [Google Scholar] [CrossRef] [PubMed]
- Kamath, A.S.; Singh, K. Perioperative Management of Pheochromocytoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: http://www.ncbi.nlm.nih.gov/books/NBK589634/ (accessed on 9 September 2025).
- Fagundes, G.F.C.; Almeida, M.Q. Perioperative Management of Pheochromocytomas and Sympathetic Paragangliomas. J. Endocr. Soc. 2022, 6, bvac004. [Google Scholar] [CrossRef]
- Taïeb, D.; Nölting, S.; Perrier, N.D.; Fassnacht, M.; Carrasquillo, J.A.; Grossman, A.B.; Clifton-Bligh, R.; Wanna, G.B.; Schwam, Z.G.; Amar, L.; et al. Management of phaeochromocytoma and paraganglioma in patients with germline SDHB pathogenic variants: An international expert Consensus statement. Nat. Rev. Endocrinol. 2024, 20, 168–184. [Google Scholar] [CrossRef] [PubMed]
- Ye, W.; Yang, W.; Guo, C.; Dong, C.; Shi, F.; Liang, C.; Ye, W.; Yang, W.; Guo, C.; Dong, C.; et al. The effect of MWA protocols upon morphology and IVIM parameters of hepatic ablation zones—A preliminary in vivo animal study with an MRI-compatible microwave ablation device. Diagn. Interv. Radiol. 2022, 28, 115–123. [Google Scholar] [CrossRef]
- Ceppa, E.P.; Collings, A.T.; Abdalla, M.; Onkendi, E.; Nelson, D.W.; Ozair, A.; Miraflor, E.; Rahman, F.; Whiteside, J.; Shah, M.M.; et al. SAGES/AHPBA Guidelines for the Use of Microwave and Radiofrequency Liver Ablation for the Surgical Treatment of Hepatocellular Carcinoma or Colorectal Liver Metastases less than 5 cm. Surg. Endosc. 2023, 37, 8991–9000. [Google Scholar] [CrossRef]
- Lubner, M.G.; Brace, C.L.; Ziemlewicz, T.J.; Hinshaw, J.L.; Lee, F.T. Microwave Ablation of Hepatic Malignancy. Semin. Intervent Radiol. 2013, 30, 56–66. [Google Scholar] [CrossRef]
- Kohlenberg, J.; Welch, B.; Hamidi, O.; Callstrom, M.; Morris, J.; Sprung, J.; Bancos, I.; Young, W. Efficacy and Safety of Ablative Therapy in the Treatment of Patients with Metastatic Pheochromocytoma and Paraganglioma. Cancers 2019, 11, 195. [Google Scholar] [CrossRef]
- Kariyasu, T.; Machida, H.; Nishina, Y.; Tambo, M.; Miyagawa, S.; Rakue, T.; Sumitani, Y.; Yasuda, K.; Shibahara, J.; Yokoyama, K. Emergent transcatheter arterial embolization to control critical blood pressure fluctuation associated with hypercatecholaminemic crisis in a patient with an unruptured retroperitoneal paraganglioma. Radiol. Case Rep. 2021, 16, 2065–2071. [Google Scholar] [CrossRef]
- Ichikawa, T.; Oyabu, C.; Minamida, M.; Ichijo, Y.; Hashimoto, Y.; Asano, M.; Iwase, H.; Tanaka, T.; Fukui, M. Changes in the Size of a Ruptured Pheochromocytoma after Transcatheter Arterial Embolization. Case Rep. Med. 2021, 2021, 5568978. [Google Scholar] [CrossRef]
- Jimenez, C.; Baudrand, R.; Uslar, T.; Bulzico, D. Perspective review: Lessons from successful clinical trials and real-world studies of systemic therapy for metastatic pheochromocytomas and paragangliomas. Ther. Adv. Med. Oncol. 2024, 16, 17588359241301359. [Google Scholar] [CrossRef] [PubMed]
- Fischer, A.; Kloos, S.; Maccio, U.; Friemel, J.; Remde, H.; Fassnacht, M.; Pamporaki, C.; Eisenhofer, G.; Timmers, H.J.L.M.; Robledo, M.; et al. Metastatic Pheochromocytoma and Paraganglioma: Somatostatin Receptor 2 Expression, Genetics, and Therapeutic Responses. J. Clin. Endocrinol. Metab. 2023, 108, 2676–2685. [Google Scholar] [CrossRef] [PubMed]
- Leijon, H.; Remes, S.; Hagström, J.; Louhimo, J.; Mäenpää, H.; Schalin-Jäntti, C.; Miettinen, M.; Haglund, C.; Arola, J. Variable somatostatin receptor subtype expression in 151 primary pheochromocytomas and paragangliomas. Hum. Pathol. 2019, 86, 66–75. [Google Scholar] [CrossRef]
- Gomes-Porras, M.; Cárdenas-Salas, J.; Álvarez-Escolá, C. Somatostatin Analogs in Clinical Practice: A Review. Int. J. Mol. Sci. 2020, 21, 1682. [Google Scholar] [CrossRef]
- Massironi, S.; Albertelli, M.; Hasballa, I.; Paravani, P.; Ferone, D.; Faggiano, A.; Danese, S. “Cold” Somatostatin Analogs in Neuroendocrine Neoplasms: Decoding Mechanisms, Overcoming Resistance, and Shaping the Future of Therapy. Cells 2025, 14, 245. [Google Scholar] [CrossRef]
- Rinke, A.; Müller, H.-H.; Schade-Brittinger, C.; Klose, K.-J.; Barth, P.; Wied, M.; Mayer, C.; Aminossadati, B.; Pape, U.-F.; Bläker, M.; et al. Placebo-Controlled, Double-Blind, Prospective, Randomized Study on the Effect of Octreotide LAR in the Control of Tumor Growth in Patients With Metastatic Neuroendocrine Midgut Tumors: A Report From the PROMID Study Group. JCO 2009, 27, 4656–4663. [Google Scholar] [CrossRef]
- Caplin, M.E.; Pavel, M.; Ćwikła, J.B.; Phan, A.T.; Raderer, M.; Sedláčková, E.; Cadiot, G.; Wolin, E.M.; Capdevila, J.; Wall, L.; et al. Lanreotide in Metastatic Enteropancreatic Neuroendocrine Tumors. N. Engl. J. Med. 2014, 371, 224–233. [Google Scholar] [CrossRef]
- Fojo, A. Exploratory Phase II Study of LAnreotide in Metastatic Pheochromocytoma/PARAganglioma (LAMPARA). clinicaltrials.gov, Clinical Trial Registration NCT03946527. February 2025. Available online: https://clinicaltrials.gov/study/NCT03946527 (accessed on 9 September 2025).
- Wolin, E.M.; Jarzab, B.; Eriksson, B.; Walter, T.; Toumpanakis, C.; Morse, M.A.; Tomassetti, P.; Weber, M.M.; Fogelman, D.R.; Ramage, J.; et al. Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des. Dev. Ther. 2015, 9, 5075–5086. [Google Scholar] [CrossRef]
- Bolanowski, M.; Kałużny, M.; Witek, P.; Jawiarczyk-Przybyłowska, A. Pasireotide—A novel somatostatin receptor ligand after 20 years of use. Rev. Endocr. Metab. Disord. 2022, 23, 601–620. [Google Scholar] [CrossRef] [PubMed]
- Vyakaranam, A.R.; Crona, J.; Norlén, O.; Granberg, D.; Garske-Román, U.; Sandström, M.; Fröss-Baron, K.; Thiis-Evensen, E.; Hellman, P.; Sundin, A. Favorable Outcome in Patients with Pheochromocytoma and Paraganglioma Treated with 177Lu-DOTATATE. Cancers 2019, 11, 909. [Google Scholar] [CrossRef]
- Severi, S.; Bongiovanni, A.; Ferrara, M.; Nicolini, S.; Di Mauro, F.; Sansovini, M.; Lolli, I.; Tardelli, E.; Cittanti, C.; Di Iorio, V.; et al. Peptide receptor radionuclide therapy in patients with metastatic progressive pheochromocytoma and paraganglioma: Long-term toxicity, efficacy and prognostic biomarker data of phase II clinical trials. ESMO Open 2021, 6, 100171. [Google Scholar] [CrossRef] [PubMed]
- Kornerup, L.S.; Andreassen, M.; Knigge, U.; Arveschoug, A.K.; Poulsen, P.L.; Kjær, A.; Oturai, P.S.; Grønbæk, H.; Dam, G. Effects of Peptide Receptor Radiotherapy in Patients with Advanced Paraganglioma and Pheochromocytoma: A Nation-Wide Cohort Study. Cancers 2024, 16, 1349. [Google Scholar] [CrossRef]
- Gubbi, S.; Al-Jundi, M.; Auh, S.; Jha, A.; Zou, J.; Shamis, I.; Meuter, L.; Knue, M.; Turkbey, B.; Lindenberg, L.; et al. Early short-term effects on catecholamine levels and pituitary function in patients with pheochromocytoma or paraganglioma treated with [177Lu]Lu-DOTA-TATE therapy. Front. Endocrinol. 2023, 14, 1275813. [Google Scholar] [CrossRef] [PubMed]
- Duan, H.; Ferri, V.; Moradi, F.; Nguyen, J.; Franc, B.L.; Iagaru, A.; Aparici, M. Peptide Receptor Radionuclide Therapy (PRRT) in Advanced Pheochromocytoma and Paraganglioma From a Single Institution Experience. 2021. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7927907/ (accessed on 17 November 2025).
- Lin, F.I.; del Rivero, J.; Carrasquillo, J.A.; Jha, A.; Zou, J.; Shamis, I.; Talvacchio, S.; Turkbey, B.; Huang, E.P.; Shih, J.; et al. Phase II Study of 177Lu-DOTATATE for Progressive Metastatic Pheochromocytomas and Paragangliomas: Interim Analysis of Efficacy, Safety, and Biomarkers. J. Clin. Oncol. 2025, 43, 3102–3112. [Google Scholar] [CrossRef]
- Tsang, E.S.; Funk, G.; Leung, J.; Kalish, G.; Kennecke, H.F. Supportive Management of Patients with Advanced Pheochromocytomas and Paragangliomas Receiving PRRT. Curr. Oncol. 2021, 28, 2823–2829. [Google Scholar] [CrossRef]
- Dillon, J.S.; Bushnell, D.; Laux, D.E. High-specific-activity 131iodine-metaiodobenzylguanidine for therapy of unresectable pheochromocytoma and paraganglioma. Future Oncol. 2021, 17, 1131–1141. [Google Scholar] [CrossRef]
- Pryma, D.A.; Chin, B.B.; Noto, R.B.; Dillon, J.S.; Perkins, S.; Solnes, L.; Kostakoglu, L.; Serafini, A.N.; Pampaloni, M.H.; Jensen, J.; et al. Efficacy and Safety of High-Specific-Activity 131I-MIBG Therapy in Patients with Advanced Pheochromocytoma or Paraganglioma. J. Nucl. Med. 2019, 60, 623–630. [Google Scholar] [CrossRef]
- Jimenez, C.; Chin, B.B.; Noto, R.B.; Dillon, J.S.; Solnes, L.; Stambler, N.; DiPippo, V.A.; Pryma, D.A. Biomarker response to high-specific-activity I-131 meta-iodobenzylguanidine in pheochromocytoma/paraganglioma. Endocr. Relat. Cancer 2022, 30, e220236. [Google Scholar] [CrossRef]
- Center for Drug Evaluation and Research. FDA Approves Iobenguane I 131 for Rare Adrenal Gland Tumors. FDA, August 2024. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-iobenguane-i-131-rare-adrenal-gland-tumors (accessed on 9 September 2025).
- Carrasquillo, J.A.; Pandit-Taskar, N.; Chen, C.C. I-131 Metaiodobenzylguanidine Therapy of Pheochromocytoma and Paraganglioma. Semin. Nucl. Med. 2016, 46, 203–214. [Google Scholar] [CrossRef]
- Jha, A.; Taïeb, D.; Carrasquillo, J.A.; Pryma, D.A.; Patel, M.; Millo, C.; de Herder, W.W.; Del Rivero, J.; Crona, J.; Shulkin, B.L.; et al. High-Specific-Activity-131I-MIBG versus 177Lu-DOTATATE Targeted Radionuclide Therapy for Metastatic Pheochromocytoma and Paraganglioma. Clin. Cancer Res. 2021, 27, 2989–2995. [Google Scholar] [CrossRef]
- Prado-Wohlwend, S.; del Olmo-García, M.I.; Bello-Arques, P.; Merino-Torres, J.F. [177Lu]Lu-DOTA-TATE and [131I]MIBG Phenotypic Imaging-Based Therapy in Metastatic/Inoperable Pheochromocytomas and Paragangliomas: Comparative Results in a Single Center. Front. Endocrinol. 2022, 13, 778322. [Google Scholar] [CrossRef]
- Zandee, W.T.; Feelders, R.A.; Smit Duijzentkunst, D.A.; Hofland, J.; Metselaar, R.M.; Oldenburg, R.A.; van Linge, A.; Kam, B.L.R.; Teunissen, J.J.M.; Korpershoek, E.; et al. Treatment of inoperable or metastatic paragangliomas and pheochromocytomas with peptide receptor radionuclide therapy using 177Lu-DOTATATE. Eur. J. Endocrinol. 2019, 181, 45–53. [Google Scholar] [CrossRef]
- Carrasquillo, J.A.; Chen, C.C.; Jha, A.; Pacak, K.; Pryma, D.A.; Lin, F.I. Systemic Radiopharmaceutical Therapy of Pheochromocytoma and Paraganglioma. J. Nucl. Med. 2021, 62, 1192–1199. [Google Scholar] [CrossRef]
- Ilanchezhian, M.; Jha, A.; Pacak, K.; Del Rivero, J. Emerging Treatments for Advanced/Metastatic Pheochromocytoma and Paraganglioma. Curr. Treat. Options Oncol. 2020, 21, 85. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Cui, Y.; Zhang, D.; Tong, A. Efficacy and Safety of Tyrosine Kinase Inhibitors in Patients with Metastatic Pheochromocytomas/Paragangliomas. J. Clin. Endocrinol. Metab. 2023, 108, 755–766. [Google Scholar] [CrossRef] [PubMed]
- Raymond, E.; Dahan, L.; Raoul, J.-L.; Bang, Y.-J.; Borbath, I.; Lombard-Bohas, C.; Valle, J.; Metrakos, P.; Smith, D.; Vinik, A.; et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011, 364, 501–513. [Google Scholar] [CrossRef] [PubMed]
- Baudin, E.; Goichot, B.; Berruti, A.; Hadoux, J.; Moalla, S.; Laboureau, S.; Nölting, S.; de la Fouchardière, C.; Kienitz, T.; Deutschbein, T.; et al. Sunitinib for metastatic progressive phaeochromocytomas and paragangliomas: Results from FIRSTMAPPP, an academic, multicentre, international, randomised, placebo-controlled, double-blind, phase 2 trial. Lancet 2024, 403, 1061–1070. [Google Scholar] [CrossRef]
- Ayala-Ramirez, M.; Chougnet, C.N.; Habra, M.A.; Palmer, J.L.; Leboulleux, S.; Cabanillas, M.E.; Caramella, C.; Anderson, P.; Al Ghuzlan, A.; Waguespack, S.G.; et al. Treatment with sunitinib for patients with progressive metastatic pheochromocytomas and sympathetic paragangliomas. J. Clin. Endocrinol. Metab. 2012, 97, 4040–4050. [Google Scholar] [CrossRef]
- Deschler-Baier, B.; Konda, B.; Massarelli, E.; Hu, M.I.; Wirth, L.J.; Xu, X.; Wright, J.; Clifton-Bligh, R.J. Clinical Activity of Selpercatinib in RET-mutant Pheochromocytoma. J. Clin. Endocrinol. Metab. 2024, 110, e600–e606. [Google Scholar] [CrossRef]
- Burotto Pichun, M.E.; Edgerly, M.; Velarde, M.; Bates, S.E.; Daerr, R.; Adams, K.; Pacak, K.; Fojo, T. Phase II clinical trial of axitinib in metastatic pheochromocytomas and paraganlgiomas (P/PG): Preliminary results. J. Clin. Oncol. 2015, 33, 457. [Google Scholar] [CrossRef]
- Jimenez, C.; Habra, M.A.; Campbell, M.T.; Tamsen, G.; Cruz-Goldberg, D.; Long, J.; Bassett, R.; Dantzer, R.; Balderrama-Brondani, V.; Varghese, J.; et al. Cabozantinib in patients with unresectable and progressive metastatic phaeochromocytoma or paraganglioma (the Natalie Trial): A single-arm, phase 2 trial. Lancet Oncol. 2024, 25, 658–667. [Google Scholar] [CrossRef]
- Tian, R.; Yao, X.; Song, J.; Wang, J.; Fu, J.; Shi, L.; Yu, F.; Zhang, P.; Zhang, C.; Ni, Y.; et al. Anlotinib for Metastatic Progressed Pheochromocytoma and Paraganglioma: A Retrospective Study of Real-World Data. J. Endocr. Soc. 2024, 8, bvae061. [Google Scholar] [CrossRef]
- Keiser, H.R.; Goldstein, D.S.; Wade, J.L.; Douglas, F.L.; Averbuch, S.D. Treatment of malignant pheochromocytoma with combination chemotherapy. Hypertension 1985, 7, I18–I24. [Google Scholar] [CrossRef]
- Averbuch, S.D.; Steakley, C.S.; Young, R.C.; Gelmann, E.P.; Goldstein, D.S.; Stull, R.; Keiser, H.R. Malignant pheochromocytoma: Effective treatment with a combination of cyclophosphamide, vincristine, and dacarbazine. Ann. Intern. Med. 1988, 109, 267–273. [Google Scholar] [CrossRef]
- Huang, H.; Abraham, J.; Hung, E.; Averbuch, S.; Merino, M.; Steinberg, S.M.; Pacak, K.; Fojo, T. Treatment of malignant pheochromocytoma/paraganglioma with cyclophosphamide, vincristine, and dacarbazine: Recommendation from a 22-year follow-up of 18 patients. Cancer 2008, 113, 2020–2028. [Google Scholar] [CrossRef] [PubMed]
- Niemeijer, N.D.; Alblas, G.; van Hulsteijn, L.T.; Dekkers, O.M.; Corssmit, E.P.M. Chemotherapy with cyclophosphamide, vincristine and dacarbazine for malignant paraganglioma and pheochromocytoma: Systematic review and meta-analysis. Clin. Endocrinol. 2014, 81, 642–651. [Google Scholar] [CrossRef]
- Asai, S.; Katabami, T.; Tsuiki, M.; Tanaka, Y.; Naruse, M. Controlling tumor progression with cyclophosphamide, vincristine, and dacarbazine treatment improves survival in patients with metastatic and unresectable malignant pheochromocytomas/paragangliomas. Horm. Cancer 2017, 8, 108–118. [Google Scholar] [CrossRef]
- Kulke, M.H.; Stuart, K.; Enzinger, P.C.; Ryan, D.P.; Clark, J.W.; Muzikansky, A.; Vincitore, M.; Michelini, A.; Fuchs, C.S. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J. Clin. Oncol. 2006, 24, 401–406. [Google Scholar] [CrossRef] [PubMed]
- Hadoux, J.; Favier, J.; Scoazec, J.-Y.; Leboulleux, S.; Al Ghuzlan, A.; Caramella, C.; Déandreis, D.; Borget, I.; Loriot, C.; Chougnet, C.; et al. SDHB mutations are associated with response to temozolomide in patients with metastatic pheochromocytoma or paraganglioma. Int. J. Cancer 2014, 135, 2711–2720. [Google Scholar] [CrossRef] [PubMed]
- Perez, K.; Jacene, H.; Hornick, J.L.; Ma, C.; Vaz, N.; Brais, L.K.; Alexander, H.; Baddoo, W.; Astone, K.; Esplin, E.D.; et al. SDHx mutations and temozolomide in malignant pheochromocytoma and paraganglioma. Endocr.-Relat. Cancer 2022, 29, 533–544. [Google Scholar] [CrossRef]
- Urquhart, C.; Fleming, B.; Harper, I.; Aloj, L.; Armstrong, R.; Hook, L.; Long, A.-M.; Jackson, C.; Gallagher, F.A.; McLean, M.A.; et al. The use of temozolomide in paediatric metastatic phaeochromocytoma/paraganglioma: A case report and literature review. Front. Endocrinol. 2022, 13, 1066208. [Google Scholar] [CrossRef] [PubMed]
- Jimenez, C.; Subbiah, V.; Stephen, B.; Ma, J.; Milton, D.; Xu, M.; Zarifa, A.; Akhmedzhanov, F.O.; Tsimberidou, A.; Habra, M.A.; et al. Phase II Clinical Trial of Pembrolizumab in Patients with Progressive Metastatic Pheochromocytomas and Paragangliomas. Cancers 2020, 12, 2307. [Google Scholar] [CrossRef]
- National Cancer Institute (NCI). DART: Dual Anti-CTLA-4 and Anti-PD-1 Blockade in Rare Tumors. clinicaltrials.gov, Clinical trial registration NCT02834013, September 2025. Available online: https://clinicaltrials.gov/study/NCT02834013 (accessed on 9 September 2025).
- Economides, M.P.; Shah, A.Y.; Jimenez, C.; Habra, M.A.; Desai, M.; Campbell, M.T. A Durable Response With the Combination of Nivolumab and Cabozantinib in a Patient With Metastatic Paraganglioma: A Case Report and Review of the Current Literature. Front. Endocrinol. 2020, 11, 594264. [Google Scholar] [CrossRef]
- Hadrava Vanova, K.; Uher, O.; Meuter, L.; Ghosal, S.; Talvacchio, S.; Patel, M.; Neuzil, J.; Pacak, K. PD-L1 expression and association with genetic background in pheochromocytoma and paraganglioma. Front. Oncol. 2022, 12, 1045517. [Google Scholar] [CrossRef]
- Yu, A.; Xu, X.; Pang, Y.; Li, M.; Luo, J.; Wang, J.; Liu, L. PD-L1 Expression is Linked to Tumor-Infiltrating T-Cell Exhaustion and Adverse Pathological Behavior in Pheochromocytoma/Paraganglioma. Lab. Investig. 2023, 103, 100210. [Google Scholar] [CrossRef]
- Celada, L.; Cubiella, T.; San-Juan-Guardado, J.; Gutiérrez, G.; Beiguela, B.; Rodriguez, R.; Poch, M.; Astudillo, A.; Grijalba, A.; Sánchez-Sobrino, P.; et al. Pseudohypoxia in paraganglioma and pheochromocytoma is associated with an immunosuppressive phenotype. J. Pathol. 2023, 259, 103–114. [Google Scholar] [CrossRef] [PubMed]
- Center for Drug Evaluation and Research. FDA Approves Belzutifan for Pheochromocytoma or Paraganglioma. FDA, May 2025. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-belzutifan-pheochromocytoma-or-paraganglioma (accessed on 9 September 2025).
- Jimenez, C.; Andreassen, M.; Durand, A.; Moog, S.; Hendifar, A.; Welin, S.; Spada, F.; Sharma, R.; Wolin, E.; Ruether, J.; et al. Belzutifan for Advanced Pheochromocytoma or Paraganglioma. N. Engl. J. Med. 2025, NEJMoa2504964. Available online: https://www.nejm.org/doi/abs/10.1056/NEJMoa2504964 (accessed on 17 November 2025). [CrossRef] [PubMed]
- FDA Grants Priority Review to Merck’s Application for WELIREG® (Belzutifan) for the Treatment of Patients with Advanced Pheochromocytoma and Paraganglioma (PPGL), Merck.com. Available online: https://www.merck.com/news/fda-grants-priority-review-to-mercks-application-for-welireg-belzutifan-for-the-treatment-of-patients-with-advanced-pheochromocytoma-and-paraganglioma-ppgl/ (accessed on 10 September 2025).
- Moog, S.; Salgues, B.; Braik-Djellas, Y.; Viel, T.; Balvay, D.; Autret, G.; Robidel, E.; Gimenez-Roqueplo, A.-P.; Tavitian, B.; Lussey-Lepoutre, C.; et al. Preclinical evaluation of targeted therapies in Sdhb-mutated tumors. Endocr.-Relat. Cancer 2022, 29, 375–388. [Google Scholar] [CrossRef]
- Hadrava Vanova, K.; Yang, C.; Meuter, L.; Neuzil, J.; Pacak, K. Reactive Oxygen Species: A Promising Therapeutic Target for SDHx-Mutated Pheochromocytoma and Paraganglioma. Cancers 2021, 13, 3769. [Google Scholar] [CrossRef]
- Enterome. A Phase 1/2 Trial of a Novel Therapeutic Vaccine (EO2401) in Combination with Immune Check Point Blockade, for Treatment of Patients with Locally Advanced or Metastatic Adrenocortical Carcinoma, or Malignant Pheochromocytoma/Paraganglioma. clinicaltrials.gov, Clinical Trial Registration NCT04187404, November 2024. Available online: https://clinicaltrials.gov/study/NCT04187404 (accessed on 10 September 2025).
- Baudin, E.; Grisanti, S.; Fassnacht, M.; C.W. der Houven van Oordt, M.-V.; Haak, H.; de la Fouchardiere, C.; Subbiah, V.; Jimenez, C.; Castillon, J.C.; Granberg, D.; et al. 2MO EO2401 (EO) therapeutic vaccine for patients (pts) with adrenocortical carcinoma (ACC) and malignant pheochromocytoma/paraganglioma (MPP): Phase I/II SPENCER study. Ann. Oncol. 2022, 33, S545–S546. [Google Scholar] [CrossRef]
- University College, London. A Phase I Trial of Vandetanib Combined with 131I-mIBG Radiotherapy in Patients with Neuroendocrine Tumours, Advanced Phaeochromocytoma and Paraganglioma. clinicaltrials.gov, Clinical TRIAL REGIStration NCT01941849, December 2015. Available online: https://clinicaltrials.gov/study/NCT01941849 (accessed on 10 September 2025).
- National Cancer Institute (NCI). A Prospective, Multi-Institutional Phase II Trial Evaluating Temozolomide vs. Temozolomide and Olaparib for Advanced Pheochromocytoma and Paraganglioma. clinicaltrials.gov, Clinical Trial Registration NCT04394858, June 2025. Available online: https://clinicaltrials.gov/study/NCT04394858 (accessed on 10 September 2025).
- Ambrosini, V.; Kunikowska, J.; Baudin, E.; Bodei, L.; Bouvier, C.; Capdevila, J.; Cremonesi, M.; de Herder, W.W.; Dromain, C.; Falconi, M.; et al. Consensus on Molecular Imaging and Theranostics in Neuroendocrine Neoplasms. Eur. J. Cancer 2021, 146, 56–73. [Google Scholar] [CrossRef]
- Nguyen, C.B.; Oh, E.; Bahar, P.; Vaishampayan, U.N.; Else, T.; Alva, A.S. Novel Approaches with HIF-2α Targeted Therapies in Metastatic Renal Cell Carcinoma. Cancers 2024, 16, 601. [Google Scholar] [CrossRef] [PubMed]
- Lantheus to Discontinue Production of Azedra. Available online: https://snmmi.org/Web/Web/News/Articles/Lantheus-to-Discontinue-Production-of-Azedra.aspx?utm_source=chatgpt.com (accessed on 2 November 2025).
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).