Pituitary carcinoma (PC) is a rare, aggressive malignancy that comprises 0.1–0.2% of all pituitary tumors. PC is defined anatomically as a pituitary tumor that metastasizes outside the primary intrasellar location as noncontiguous lesions in the central nervous system or as metastases to other organs. Similar to pituitary adenoma, PC originates from various cell types of the pituitary gland and can be functioning or nonfunctioning, with the former constituting the majority of the cases. Compression of intricate skull-based structures, excessive hormonal secretion, impaired pituitary function from therapy, and systemic metastases lead to debilitating symptoms and a poor survival outcome in most cases. PC frequently recurs despite multimodality treatments, including surgical resection, radiotherapy, and biochemical and cytotoxic treatments. There is an unmet need to better understand the pathogenesis and molecular characterization of PC to improve therapeutic strategies. As our understanding of the role of signaling pathways in the tumorigenesis of and malignant transformation of PC evolves, efforts have focused on targeted therapy. In addition, recent advances in the use of immune checkpoint inhibitors to treat various solid cancers have led to an interest in exploring the role of immunotherapy for the treatment of aggressive refractory pituitary tumors. Here, we review our current understanding of the pathogenesis, molecular characterization, and treatment of PC. Particular attention is given to emerging treatment options, including targeted therapy, immunotherapy, and peptide receptor radionuclide therapy.

The majority of pituitary neoplasms are pituitary adenomas (PAs), which are common, benign glandular tumors that are derived from the adenohypophysis and classified by their endocrinologic cell lineage.[1] These tumors are also broadly differentiated by size as either macroadenomas (≥ 1 cm) or microadenomas (< 1 cm). Treatment of PA is indicated when patients become symptomatic, either from mass effect or from the excess production of hormones such as prolactin, adrenocorticotropic hormone (ACTH), growth hormone (GH), and thyroid-stimulating hormone (TSH).[2] Most symptomatic PAs have a favorable response to the combination of surgical resection and hormonally targeted therapies, with a 5-year survival rate of > 90%.[3] Less commonly, these tumors are locally destructive, treatment resistant, and have high recurrence rates, which are defining features of what the World Health Organization (WHO) now recognizes as aggressive pituitary tumors (APTs).[4] In the most extreme cases, 0.1–0.2% of tumors metastasize outside of their primary intrasellar location as noncontiguous foci within the central nervous system or systemically, becoming pituitary carcinomas (PCs).[57] While no well-established histopathologic, molecular, or genetic distinctions exist between PA and APT and PC,[8] the metastatic nature of PC establishes it as a clinically distinct entity.[9] To avoid the need to reclassify PAs as PCs upon the identification of metastases, the WHO proposed that pituitary tumors be classified as pituitary neuroendocrine tumors to encompass both entities.[10,11]

An adenoma-to-carcinoma progression model of PC has been hypothesized,[12] with limited evidence suggesting rare cases of de novo tumor origination.[13] Studies have reported a highly variable latency period between the diagnosis of adenoma to the first metastatic lesion, with a mean of approximately 5–6 years and occurrence as late as 29 years after the initial diagnosis.[14,15] Efforts to identify morphologic, immunohistochemical, or molecular markers to predict metastatic potential are still ongoing.[16] Greater than 70% of PCs are functional, with corticotroph (ACTH-producing) and lactotroph (prolactin-producing) tumors being the most common, followed by less frequently reported cases of luteinizing hormone, GH- or follicle-stimulating hormone, and TSH-producing tumors.[17]

The initial clinical presentation of PC mirrors that of invasive PA; most clinical manifestations result from local sellar and cavernous sinus impingement, including headaches, visual disturbances, cranial nerve palsy, and hormonal imbalances, such as Cushing disease (hypercortisolism).[18] A diagnosis cannot be made until metastatic disease is identified,[19] at which point management is increasingly difficult. Treatment strategies include surgical resection, radiotherapy, and biochemical and cytotoxic treatments. Temozolomide (TMZ), an alkylating chemotherapy agent, has shown efficacy in the treatment of PC,[20] but sustained treatment responses are uncommon, with frequent disease progression and a 2-year survival rate not exceeding 50%.[2123]

Because of the diagnostic and therapeutic challenges that arise in managing this rare entity, in this review paper we summarize our current understanding of PC by reviewing recent advances in its diagnosis, molecular characterization, and treatment, with particular attention to the emerging role of immunotherapy for patients with this rare, aggressive malignancy.

Pathogenesis and Molecular Characterization

The pathogenesis of PA and its transition to APT and PC is complex and poorly understood; elucidating a unifying paradigm has been difficult because of inherent differences among pituitary cell subtypes. Given the trophic influence of hypothalamic hormones on stimulation of the anterior pituitary gland, hypothalamic dysregulation may provide the initial proliferative stimulus that triggers pituitary cell growth. However, it seems more likely that intrapituitary factors, namely the activation of selective oncoproteins or loss of suppressor factors, are the primary drivers of tumorigenesis.[24]

In one study, deregulation in the Rb/p16/cyclin D1/cyclin–dependent kinase 4 pathway was found in up to 80% of PAs.[25] The pituitary tumor–transforming gene, a regulator of anaphase and activator of growth signals that include vascular endothelial growth factor (VEGF)/fibroblast growth factor,[26] was found to be overexpressed in 90% of adenomas, with little or no expression in healthy pituitary tissue.[27,28] It has also been theorized that the benign nature of most PAs may result partially from oncogene-induced senescence,[29,30] a mechanism in which early genomic aberrations, including alterations in the pituitary tumor–transforming gene, trigger powerful intrinsic tumor-suppressive activities, mainly through the p53/p21 and p16/Rb inhibitory pathways.[31] Experimental pituitary tumor models in rats have demonstrated increasing levels of nuclear p21 over time after an initial proliferative phase in somatolactotroph cell lines.[32] An immunohistochemical study also showed lower nuclear staining of p16 in all pituitary tumors than that in normal pituitary tissue, with the decrease most pronounced in six cases of PC.[33  The loss of oncogene-induced senescence mechanisms seems to be a recurrent factor in the malignant transformation observed in PCs.[34] These findings are accompanied by the caveat that these and other senescence patterns are not consistent across pituitary cell subtypes.[35]

In addition, the upregulation of proangiogenic factors, most notably including epidermal growth factor, VEGF, and matrix metalloproteinase-9, have been implicated in PC pathogenesis.[36] Numerous other mediators, such as the upregulation of cyclin-dependent kinases and downregulation of apoptotic proteins such as Bcl-2, have also been implicated as potential contributors to aggressive behavior and recurrence.[33] It remains challenging to identify key molecular patterns within the malignant transformation of these heterogeneous tumors because the evolution from adenoma to carcinoma is typically gradual, with an extended clinical latency.

Currently, pituitary tumors are characterized mainly by proliferative markers, with the WHO recommending the use of Ki-67 index > 3%, p53 immunoreactivity, and an elevated mitotic count as potential markers of clinical aggressiveness.[37] The validity of Ki-67 as a prognostic marker is recognized in many tumors,[38] but its use remains unclear in pituitary tumors.[39] Multiple studies refute its prognostic value,[40] while others demonstrate a significant inverse relationship between the Ki-67 index and the risk of recurrence,[41] as well as the ability to stratify among noninvasive adenomas, APTs, and PCs on the basis of the degree of Ki-67 index elevation.[42] Ultimately, while the established cutoff of 3% is controversial, Ki-67 seems to have high sensitivity but low specificity for predicting tumor recurrence and invasive potential.[43] Thus, other immunohistochemical markers, such as p53, and other clinical characteristics, such as resistance to conventional treatment regimens and regrowth after repeat resections, should also be used to prompt more aggressive early management and more frequent interval imaging and endocrine evaluations.[44]

PAs become clinically significant and prompt treatment as a result of excess hormone production or mass effect and impingement on surrounding cranial structures.[45] The initial course may involve medical therapy, as is the case with prolactinomas, for which dopamine agonists such as bromocriptine or cabergoline are often sufficient to reduce tumor size and achieve biochemical remission, or primary surgical resection in the case of ACTH-, GH-, or TSH-producing adenomas.[46] Surgical approaches emphasize maximal resection, with a preferred transsphenoidal approach, to alleviate mass effect and facilitate biochemical remission.[47] Radiotherapy is generally used when surgery is inadequate to control the tumor or in the setting of inoperable recurrence.[48] Stereotactic radiosurgery is generally preferred,[49] although when targeting lesions near or within the optic chiasm, fractionated radiotherapy is often used to reduce the risk of developing an optic neuropathy.[50]

Medical therapy for PA and APT involves reducing excess hormone secretion from functional pituitary tumors using medications such as somatostatin analogs (octreotide, lanreotide, and pasireotide) and dopamine agonists (bromocriptine and cabergoline). Somatostatin analogs exert counterregulatory effects on the release of a variety of hormones, including GH, TSH, and ACTH. Bromocriptine and cabergoline act as agonists at pituitary D2 receptors, inhibiting the release of GH, prolactin, and ACTH by increasing dopamine antagonism.

The initial treatment of PCs involves multidisciplinary assessment for re-resection, reirradiation, hormonal therapies, and cytotoxic chemotherapies. Of note, reirradiation has been shown to be helpful in controlling tumor mass but not necessarily in limiting excess humoral secretion from these tumors.[51] Thus far, TMZ, an alkylating agent that is used as the standard of care for high-grade gliomas, has shown the most promise and is now established as a first-line chemotherapy option for the treatment of PCs.[52] In 2018, the European Society of Endocrinology published clinical practice guidelines that included a recommendation for TMZ monotherapy after the failure of standard therapies, using standard 150–200 mg/m2 dosing for 5 days every 28 days, treatment evaluation after three cycles, and the continuation of therapy for at least 6 months in patients who experienced a response to this initial three-cycle regimen.[44] Several recent larger-scale retrospective studies have noted median progression-free survival (mPFS) durations ranging from 23 to 40 months with TMZ monotherapy.[53,54] In a recent large-scale meta-analysis of 21 studies involving 421 patients with either APTs or PCs, an mPFS duration of 20 months was noted with a 40% radiologic response rate in the patient cohort, which improved to 60% with chemoradiotherapy.[55] While there are cases that demonstrate more durable, sustained treatment responses,[56,57] a substantial proportion of patients with PC do not experience a response to TMZ or experience recurrence after an initial response.

Given these findings, further research has sought to understand the potential predictors of response to TMZ therapy in APTs and PCs. Previous studies have established that low expression of O6-methylguanine-DNA methyltransferase (MGMT), a DNA repair enzyme that mechanistically counteracts the alkylating effects of TMZ, is associated with chemoresistance in various gliomas.[58] In one study of 24 patients with PCs, the tumor cell nuclei of nonresponders had median MGMT staining of 93% compared with 9% in responders.[59] An independent research group also reported an association between low MGMT expression and positive treatment response in PCs.[60] However, other studies have suggested a more equivocal relationship, which is likely complicated by conflicting interstudy reliability and inconsistent immunohistochemistry staining methods.[61] Overall, the evidence is not robust enough to exclude from therapy patients who have high MGMT expression; however, the previously mentioned European Society of Endocrinology clinical guidelines still recommend determining MGMT status via immunohistochemistry as a tool to guide therapy (low-evidence recommendation).[44] Another DNA repair gene that is implicated in the prediction of response to TMZ is MSH6, a gene that codes for a DNA mismatch repair protein for which mutations are associated with an increased risk of malignancy, such as that seen in Lynch syndrome. One case reported the evolution of TMZ resistance in a patient with PC and an MSH6 mutation from immunopositive to immunonegative in the setting of an otherwise MGMT-negative, p53-mutated lactotroph carcinoma, suggesting that loss of MSH6 was the driver of resistance to TMZ.[62] A small-scale retrospective analysis of 13 patients, 10 of whom were noted to have PC, demonstrated that immunopositivity of MSH6, but not Ki-67, p53, or MGMT, was correlated with an improved response to TMZ, with a lower likelihood of progressive disease.[63]

Given the high rates of relapse after TMZ monotherapy, there have been efforts to use combinatorial approaches to treat PC. In one case series, the use of concurrent radiotherapy and TMZ resulted in the sustained control of treatment-resistant extraneural metastases in two patients with PC.[64] The addition of capecitabine to TMZ (CAPTEM) is another regimen that has been shown to be effective. Capecitabine is a prodrug of 5-FU, an antimetabolite agent that has been shown to be synergistic with TMZ in vivo in the treatment of neuroendocrine neoplasms.[65] In a large clinical trial of 144 patients with advanced pancreatic neuroendocrine tumors, the mPFS duration was 22.7 months in patients receiving CAPTEM compared to 14.4 months in patients receiving TMZ alone.[66] In one case series of four patients with corticotroph PCs, two patients experienced complete disease regression, and one had stable disease for > 4.5 years after CAPTEM.[67] However, other available case reports have demonstrated more variable responses[68,69]; ultimately, prospective trials will be needed to elucidate whether CAPTEM is superior to TMZ monotherapy in patients with PC. In cases of TMZ nonresponse, case reports have demonstrated varying degrees of success for salvage regimens, including etoposide with cisplatin/carboplatin.[7072] In one case of a patient diagnosed with a corticotroph carcinoma at age 14, treatment with carboplatin and 5-fluorouracil resulted in prolonged survival; the patient is noted to still be in disease remission over a decade after her initial diagnosis.[73]

Targeted Therapies

As our understanding of the role of angiogenic growth factors and the PI3KAkt//mTOR pathway on the tumorigenesis of malignant endocrine tumors evolves,[74] efforts have focused on targeted therapy as an alternative strategy to improve clinical outcomes in APT and PC.

The expression of VEGF is upregulated in both invasive adenoma and PC compared to noninvasive adenoma,[75,76] suggesting a role for targeting VEGF with bevacizumab or the VEGF receptor with sunitinib or sorafenib.[77] One case report demonstrated a lack of disease progression on bevacizumab for 26 months after TMZ failure,[78] and another demonstrated 5 years of stability when bevacizumab was combined with TMZ.[79] A case treated with bevacizumab following disease progression on checkpoint inhibitor (CPI) therapy resulted in an 8-month progression-free (PFS) survival.[80] Lapatinib, a tyrosine kinase inhibitor that targets epidermal growth factor has been approved for use in metastatic HER2 breast cancer; it was also tested in a recent phase 2 prospective trial of treatment-resistant prolactinoma and was effective in three cases of locally invasive APT, but not in a patient with prolactinoma with craniospinal metastasis.[81] In contrast to anti-VEGF and epidermal growth factor therapies, targeting mTOR through the use of everolimus has thus far not been effective in PC when used as monotherapy[82] or in combination with hormonal therapies.[83] It seems plausible that these and other targeted therapies are more effective in carefully selected patients with more activating mutations, although currently there is no specific evidence to validate this hypothesis. While current evidence regarding targeted therapy use in patients with PC and APTs is limited, there is some preliminary evidence that combinatorial approaches using targeted and cytotoxic therapies can be effective in a subset of patients.

Immunotherapy

The immune tumor microenvironment involves a complex interplay among many different tissue components, including infiltrating immune cells, tumor cells, resident tissue cells, such as fibroblasts and endothelial cells, and the extracellular matrix.[84] Through these complex interactions, tumor cells evade immune responses through several mechanisms, including the upregulation of coinhibitory cytotoxic T lymphocyte–associated protein 4 (CTLA-4) and the programmed death ligand 1 and 2 (PD-L1 and PD-L2, respectively) pathways,[85] which have been shown to be present to variable degrees across pituitary tumor subtypes.[86] A recent analysis of 60 pituitary tumor samples demonstrated increased expression of PD-L2 and CD80/CD86, coreceptors that are able to interact with CTLA-4, in more APTs.[87] Furthermore, hypophysitis and hypopituitarism are well-established adverse events seen primarily in patients receiving CTLA-4 blockade.[88] Given these findings, there is a mechanistic basis for the use of immunotherapy in APT and PC.[89]

A recent phase 2 trial of pembrolizumab, a PD-L1 receptor blocker, in rare tumors included four patients with PC.[90] One patient with a corticotroph carcinoma experienced disease progression after TMZ, CAPTEM, and multiple rounds of targeted radiotherapy. However, after treatment with pembrolizumab, the patient had regression of intracranial and metastatic disease that was sustained for 42 months after treatment initiation. Interestingly, this patient was noted to have a hypermutator phenotype that included both MSH2 and MSH6 mutations. Another patient with a corticotroph carcinoma, who experienced disease progression after stereotactic radiosurgery and TMZ, also demonstrated a partial response, with a progression-free survival duration of 12 months at the time of study conclusion. The other two patients, one with silent corticotroph carcinoma and one with lactotroph carcinoma, did not experience a response to pembrolizumab.[91]

In addition to pembrolizumab monotherapy, five individual case reports have noted success with dual immune-checkpoint blockade in the treatment of PC. In one patient with functional corticotroph carcinoma with hepatic metastases who received therapy with nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4), five cycles of therapy resulted in a 92% reduction in dominant hepatic metastasis and a 59% reduction in the recurrent intracranial component, along with stable disease 6 months after study conclusion on nivolumab maintenance.[92] In one case of corticotroph carcinoma, stable disease was observed 1 year after ipilimumab and nivolumab therapy,[93] while two other cases reported 8 months of stable disease on this regimen.[80,94] Recently, a patient with lactotroph carcinoma treated with ipilimumab and nivolumab had complete, sustained remission 24 months after the initiation of therapy. To date and to our knowledge, this is the only reported case with complete radiologic and endocrinologic response.[95] Evidently, although a subset of patients responded to CPI therapy, larger-scale prospective trials are needed to further clarify the role of these agents. Ongoing studies (ClinicalTrials.gov Identifiers: NCT02834013 and NCT04042753) are testing the efficacy of ipilimumab and nivolumab in APT and PC patients.[89] A summary of the results of key immunotherapy trials conducted in patients with pituitary carcinoma can be referenced in Table 1.

A recent systematic review of the use of immunotherapy specifically in APTs and PCs noted several important trends in the observed cases, including the slightly more favorable therapeutic response in corticotroph than in lactotroph carcinoma.[96] This may be the result of an increased number of infiltrating CD8+ T cells in functional corticotroph tumors compared with lactotroph and other tumor subtypes.[97] While such a proposed mechanism is plausible, there remains a need to determine whether the biomarkers used to predict treatment response of CPI in other solid tumor malignancies (such as tumor-mutational burden [TMB], microsatellite instability, PD-L1, and increased tumor-infiltrating T cells) are also predictive of treatment response in APTs and PCs.[98]

In particular, TMB, defined as the number of somatic mutations per coding area of a tumor's genome, has been an emerging clinical biomarker[99]; hypothetically, tumors with high TMB have an increased number of neoantigens, which can help native immune cells to recognize and kill tumor cells.[100] TMB could be of particular relevance to PC because of the high rate of hypermutation that occurs commonly after TMZ use, and the poor prognosis, such hypermutation, typically portends.[101] However, the correlation of high TMB with response to immunotherapy has been demonstrated to be tumor specific, showing favorable prognostic value in melanoma and lung but not in gliomas, for example.[102] Given demonstrated tumor-specific correlation, future studies must ascertain whether high TMB could be useful as a predictor of response to immune checkpoint inhibitor (ICI) therapy. Based on a recent retrospective, observational cohort study that included 15 PCs and APTs treated with ICIs in France, four corticotroph carcinomas with negative PDL-1 staining and < 1% CD8+ T cell infiltration demonstrated partial response. This result suggests that the lack of presence of these typically favorable prognostic markers does not preclude immunotherapy response[103]; however, neither high TMB nor microsatellite instability have been specifically studied yet.

Peptide Receptor Radionuclide Therapy

Pituitary tumor histotypes have been shown to express, widely and heterogeneously, the various somatostatin receptor (SSTR) subtypes, with somatotrophs, for example, mainly expressing SSTR2 and SSTR5 and lactotrophs expressing SST1 and SST5.[104] Peptide receptor radionuclide therapy (PRRT) technology uses radiolabeled peptides, including somatostatin analogues such as 177Lu DOTA-TATE, to target SSTR receptors and selectively deliver cytotoxic doses of radiation.[105]

PRRT has been evaluated to a limited extent in APTs and PCs. Of 13 total published cases of PRRT in patients with APT in the medical literature, four experienced a clinical response, defined by either growth arrest or shrinkage of the tumor bulk, improvement in clinical signs and symptoms, or biochemical improvement.[106] In our experience, one patient with a metastatic prolactinoma progressed after a single course of PRRT. To date, there have been three reported cases of success with PRRT in PC. One patient demonstrated stable disease for over 4 years after the initiation of treatment,[107] and another remained radiologically stable, with a complete response in some leptomeningeal nodules 40 months after treatment induction.[108] A more recent case report of a patient with highly resistant corticotroph PC demonstrated sustained clinical stability of over a year after four doses of Lu DOTA-TATE were given following ipilumumab and nivolumab, suggesting a synergistic response between PRRT and CPI.[109] A notable potential advantage of PRRT is its eventual ability to effectively differentiate between clinical responders and nonresponders through the use of quantitative positron emission tomography (PET)-derived parameters on SSTR imaging.[110] The use of pretherapeutic maximum standardized uptake values (SUVmax) of various radiolabeled somatostatin analogs on PET/CT scans have been demonstrated to both be correlated strongly with PRRT response[111] and be predictive of PRRT, with greater than 95% sensitivity or specificity in some studies.[112]

Conversely, a limitation of PRRT is its toxicity, with marrow suppression and nephrotoxicity being major dose-limiting side effects,[113] a particular concern for PC patients who have already received cytotoxic chemotherapy. In one larger-scale retrospective analysis of 807 patients who underwent PRRT for neuroendocrine tumors, 2.5% of patients developed myelodysplastic syndrome; 33% of patients also experienced some degree of nephrotoxicity, although it was severe (grade 3–4) in only 1%.[105]

Of note, there is currently a phase 2 clinical trial for PRRT use in patients with SSTR+ neuroendocrine tumors for which patients with PC would be eligible, and the trial will help clinicians to further determine the efficacy for PRRT use in patients with PC.

PC is a rare neuroendocrine malignancy that remains challenging to treat given that no well-defined standard therapy exists. Treatment recommendations are largely driven by small prospective and retrospective case series. Most patients undergo resection, focal radiotherapy, and medical therapy that is directed at reducing hormone hypersecretion and its clinical manifestations (biochemical therapy), decreasing tumor size to improve mass effect and related neurologic symptoms (chemotherapy), and correcting hormone deficiencies. TMZ remains the first-line chemotherapy to treat PC, which is refractory to the above standard therapies. Other combinatorial approaches, such as CAPTEM and therapies targeting VEGF and CPI, seem to offer the greatest potential to improve outcomes. Conducting clinical trials in PC is exceedingly difficult because of the rarity of the disease and the lack of access to specialized centers by all patients. Therefore, large multicenter clinical trial efforts are needed to be able to conduct meaningful research in this orphan disease. Importantly, future research should focus on establishing a national tumor bank and databases for APTs and PCs to expand our understanding of their molecular pathogenesis and resistance to different treatment modalities.

1.
Melmed
S.
Pituitary tumors
.
Endocrinol Metab Clin North Am
.
2015
;
44
:
1
9
.
2.
Neggers
SJCMM,
van der Lely
AJ.
Medical approach to pituitary tumors
.
In:
Handbook of Clinical Neurology
.
Vol 124. Elsevier;
2014
:
303
316
.
3.
Chen
C,
Hu
Y,
Lyu
L,
et al
Incidence, demographics, and survival of patients with primary pituitary tumors: a SEER database study in 2004–2016
.
Sci Rep
.
2021
;
11
:
15155
.
4.
Mete
O,
Lopes
MB.
Overview of the 2017 WHO classification of pituitary tumors
.
Endocr Pathol
.
2017
;
28
:
228
243
.
5.
Dekkers
OM,
Karavitaki
N,
Pereira
AM.
The epidemiology of aggressive pituitary tumors (and its challenges)
.
Rev Endocr Metab Disord
.
2020
;
21
:
209
212
.
6.
Kaltsas
GA,
Nomikos
P,
Kontogeorgos
G,
et al
Clinical review: diagnosis and management of pituitary carcinomas
.
J Clin Endocrinol Metab
.
2005
;
90
:
3089
3099
.
7.
Heaney
AP.
Clinical review: pituitary carcinoma: difficult diagnosis and treatment
.
J Clin Endocrinol Metab
.
2011
;
96
:
3649
3660
.
8.
Chatzellis
E,
Alexandraki
KI,
Androulakis
Kaltsas G.
Aggressive pituitary tumors
.
Neuroendocrinology
.
2015
;
101
:
87
104
.
9.
Raverot
G,
Ilie
MD,
Lasolle
H,
et al
Aggressive pituitary tumours and pituitary carcinomas
.
Nat Rev Endocrinol
.
2021
;
17
:
671
684
.
10.
Rindi
G,
Klimstra
DS,
Abedi-Ardekani
B,
et al
A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal
.
Mod Pathol
.
2018
;
31
:
1770
1786
.
11.
Louis
DN,
Perry
A,
Wesseling
P,
et al
The 2021 WHO classification of tumors of the central nervous system: a summary
.
Neuro Oncol
.
2021
;
23
:
1231
1251
.
12.
Xu
L,
Khaddour
K,
Chen
J,
Rich
KM,
Perrin
RJ,
Campian
JL.
Pituitary carcinoma: two case reports and review of literature
.
World J Clin Oncol
.
2020
;
11
:
91
102
.
13.
Scheithauer
BW,
Kovacs
K,
Nose
V,
et al
Multiple endocrine neoplasia type 1–associated thyrotropin-producing pituitary carcinoma: report of a probable de novo example
.
Hum Pathol
.
2009
;
40
:
270
278
.
14.
Santos-Pinheiro
F,
Penas-Prado
M,
Kamiya-Matsuoka
C,
et al
Treatment and long-term outcomes in pituitary carcinoma: a cohort study
.
Eur J Endocrinol
.
2019
;
181
:
397
407
.
15.
Wang
J,
Ma
EM,
Wu
PF,
et al
Multiple intracranial and spinal metastases from a nonfunctioning pituitary adenoma following multiple surgeries: an illustrative case with 16 years of follow-up
.
World J Surg Oncol
.
2014
;
12
:
380
.
16.
Alshaikh
OM,
Asa
SL,
Mete
O,
Ezzat
S.
An institutional experience of tumor progression to pituitary carcinoma in a 15-year cohort of 1055 consecutive pituitary neuroendocrine tumors
.
Endocr Pathol
.
2019
;
30
:
118
127
.
17.
Ragel
BT,
Couldwell
WT.
Pituitary carcinoma: a review of the literature
.
Neurosurg Focus
.
2004
;
16
:
1
9
.
18.
Brue
T,
Castinetti
F.
The risks of overlooking the diagnosis of secreting pituitary adenomas
.
Orphanet J Rare Dis
.
2016
;
11
:
135
.
19.
Lenders
N,
McCormack
A.
Malignant transformation in non-functioning pituitary adenomas (pituitary carcinoma)
.
Pituitary
.
2018
;
21
:
217
229
.
20.
Ji
Y,
Vogel
RI,
Lou
E.
Temozolomide treatment of pituitary carcinomas and atypical adenomas: systematic review of case reports
.
Neurooncol Pract
.
2016
;
3
:
188
195
.
21.
Shen
AJ,
King
J,
Colman
PG,
Yates
CJ.
Diagnosis and management of adrenocorticotropic hormone-secreting pituitary carcinoma: a case report and review of the literature. Future Rare Dis.
2021
;
1:FRD13.
22.
Yoo
F,
Kuan
EC,
Heaney
AP,
et al
Corticotrophic pituitary carcinoma with cervical metastases: case series and literature review
.
Pituitary
.
2018
;
21
:
290
301
.
23.
Hansen
TM,
Batra
S,
Lim
M,
et al
Invasive adenoma, and pituitary carcinoma: a SEER database analysis
.
Neurosurg Rev
.
2014
;
37
:
279
286
.
24.
Clayton
RN,
Farrell
WE.
Clonality of pituitary tumours: more complicated than initially envisaged?
Brain Pathol
.
2001
;
11
:
313
327
.
25.
Muşat
M,
Vax
VV,
Borboli
N,
et al
Cell cycle dysregulation in pituitary oncogenesis
.
Front Horm Res
.
2004
;
32
:
34
62
.
26.
Xiong
Z,
Li
X,
Yang
Q.
PTTG has a dual role of promotion-inhibition in the development of pituitary adenomas
.
Protein Pept Lett
.
2019
;
26
:
800
818
.
27.
Tfelt-Hansen
J,
Kanuparthi
D,
Chattopadhyay
N.
the emerging role of pituitary tumor transforming gene in tumorigenesis
.
Clin Med Res
.
2006
;
4
:
130
137
.
28.
Elsarrag
M,
Patel
PD,
Chatrath
A,
et al
Genomic and molecular characterization of pituitary adenoma pathogenesis: review and translational opportunities
.
Neurosurg Focus
.
2020
;
48
:
E11
.
29.
Mooi
WJ.
Oncogene-induced cellular senescence: causal factor in the growth arrest of pituitary microadenomas?
Horm Res
.
2009
;
71
:
78
81
.
30.
Zhu
H,
Blake
S,
Kusuma
FK,
et al
Oncogene-induced senescence: from biology to therapy
.
Mech Ageing Dev
.
2020
;
187
:
111229
.
31.
Chesnokova
V,
Melmed
S.
Pituitary senescence: the evolving role of Pttg
.
Mol Cell Endocrinol
.
2010
;
326
:
55
59
.
32.
Chesnokova
V,
Zonis
S,
Rubinek
T,
et al
Senescence mediates pituitary hypoplasia and restrains pituitary tumor growth
.
Cancer Res
.
2007
;
67
:
10564
10572
.
33.
Alexandraki
KI,
Munayem
Khan
M,
Chahal
HS,
et al
Oncogene-induced senescence in pituitary adenomas and carcinomas
.
Hormones (Athens)
.
2012
;
11
:
297
307
.
34.
Sav
A,
Rotondo
F,
Syro
LV,
et al
Selective molecular biomarkers to predict biologic behavior in pituitary tumors
.
Expert Rev Endocrinol Metab
.
2017
;
12
:
177
185
.
35.
Manojlovic-Gacic
E,
Skender-Gazibara
M,
Popovic
V,
et al
Oncogene-induced senescence in pituitary adenomas—an immunohistochemical study
.
Endocr Pathol
.
2016
;
27
:
1
11
.
36.
Yang
Z,
Zhang
T,
Gao
H.
Genetic aspects of pituitary carcinoma: a systematic review
.
Medicine
.
2016
;
95
:
e5268
.
37.
Nishioka
H,
Inoshita
N.
New WHO classification of pituitary adenomas (4th edition): assessment of pituitary transcription factors and the prognostic histological factors
.
Brain Tumor Pathol
.
2018
;
35
(2)
:
57
61
.
38.
Salehi
F,
Agur
A,
Scheithauer
BW,
et al
Ki-67 in pituitary neoplasms: a review—part I
.
Neurosurgery
.
2009
;
65
:
429
437
;
discussion 437.
39.
Yakoushina
TV,
Lavi
E,
Hoda
RS.
Pituitary carcinoma diagnosed on fine needle aspiration: report of a case and review of pathogenesis
.
CytoJournal
.
2010
;
7
:
14
.
40.
Grimm
F,
Maurus
R,
Beschorner
R,
et al
Ki-67 labeling index and expression of p53 are non-predictive for invasiveness and tumor size in functional and nonfunctional pituitary adenomas
.
Acta Neurochir
.
2019
;
161
:
1149
1156
.
41.
Hasanov
R,
Aydoğan
Bİ,
Kiremitçi
S,
et al
The prognostic roles of the Ki-67 proliferation index, P53 expression, mitotic index, and radiological tumor invasion in pituitary adenomas
.
Endocr Pathol
.
2019
;
30
:
49
55
.
42.
Thapar
K,
Kovacs
K,
Scheithauer
BW,
et al
Proliferative activity and invasiveness among pituitary adenomas and carcinomas: an analysis using the MIB-1 antibody
.
Neurosurgery
.
1996
;
38
:
99
106
;
discussion 106–107.
43.
Lu
C,
Liu
Y,
Lu
Z,
Huan
C.
Ki-67, and clinical correlations in patients with resistant prolactinomas
.
Ann Clin Lab Sci
.
2020
;
50
:
199
204
.
44.
Raverot
G,
Burman
P,
McCormack
A,
et al
European Society of Endocrinology Clinical Practice Guidelines for the management of aggressive pituitary tumours and carcinomas
.
Eur J Endocrinol
.
2018
;
178
:
G1
G24
.
45.
Esposito
D,
Olsson
DS,
Ragnarsson
O,
et al
Non-functioning pituitary adenomas: indications for pituitary surgery and post-surgical management
.
Pituitary
.
2019
;
22
:
422
434
.
46.
Molitch
ME.
Diagnosis and treatment of pituitary adenomas: a review
.
JAMA
.
2017
;
317
:
516
.
47.
Mehta
GU,
Lonser
RR.
Management of hormone-secreting pituitary adenomas. Neuro Oncol. Published online Aug 19,
2016
.
48.
Loeffler
JS,
Shih
HA.
Radiation therapy in the management of pituitary adenomas
.
J Clin Endocrinol Metab
.
2011
;
96
:
1992
2003
.
49.
Landolt
AM,
Haller
D,
Lomax
N,
et al
Stereotactic radiosurgery for recurrent surgically treated acromegaly: comparison with fractionated radiotherapy
.
J Neurosurg
.
1998
;
88
:
1002
1008
.
50.
Tishler
RB,
Loeffler
JS,
Lunsford
LD,
et al
Tolerance of cranial nerves of the cavernous sinus to radiosurgery
.
Int J Radiat Oncol Biol Phys
.
1993
;
27
:
215
221
.
51.
Verma
J,
McCutcheon
IE,
Waguespack
SG,
Mahajan
A.
Feasibility, and outcome of re-irradiation in the treatment of multiply recurrent pituitary adenomas
.
Pituitary
.
2014
;
17
:
539
545
.
52.
McCormack
A,
Dekkers
OM,
Petersenn
S,
et al
Treatment of aggressive pituitary tumours and carcinomas: results of a European Society of Endocrinology (ESE) survey 2016
.
Eur J Endocrinol
.
2018
;
178
:
265
276
.
53.
Elbelt
U,
Schlaffer
SM,
Buchfelder
M,
et al
Efficacy of temozolomide therapy in patients with aggressive pituitary adenomas and carcinomas–a German survey. J Clin Endocrinol Metab.
2020
;
105:dgz211.
54.
Lasolle
H,
Cortet
C,
Castinetti
F,
et al
Temozolomide treatment can improve overall survival in aggressive pituitary tumors and pituitary carcinomas
.
Eur J Endocrinol
.
2017
;
176
:
769
777
.
55.
Luo
M,
Tan
Y,
Chen
W,
et al
Clinical efficacy of temozolomide and its predictors in aggressive pituitary tumors and pituitary carcinomas: a systematic review and meta-analysis
.
Front Neurol
.
2021
;
12
:
700007
.
56.
Jordan
JT,
Miller
JJ,
Cushing
T,
et al
Temozolomide therapy for aggressive functioning pituitary adenomas refractory to surgery and radiation: a case series
.
Neurooncol Prac
.
2018
;
5
:
64
68
.
57.
Hagen
C,
Schroeder
HD,
Hansen
S,
et al
Temozolomide treatment of a pituitary carcinoma and two pituitary macroadenomas resistant to conventional therapy
.
Eur J Endocrinol
.
2009
;
161
:
631
637
.
58.
Hegi
ME,
Diserens
AC,
Gorlia
T,
et al
MGMT gene silencing and benefit from temozolomide in glioblastoma
.
N Engl J Med
.
2005
;
352
:
997
1003
.
59.
Bengtsson
D,
Schrøder
HD,
Andersen
M,
et al
Long-term outcome and MGMT as a predictive marker in 24 patients with atypical pituitary adenomas and pituitary carcinomas given treatment with temozolomide
.
J Clin Endocrinol Metab
.
2015
;
100
:
1689
1698
.
60.
Kovacs
K,
Scheithauer
BW,
Lombardero
M,
et al
MGMT immunoexpression predicts responsiveness of pituitary tumors to temozolomide therapy
.
Acta Neuropathol
.
2008
;
115
:
261
262
.
61.
Yu
W,
Zhang
L,
Wei
Q,
Shao
A.
O6-Methylguanine-DNA methyltransferase (MGMT): challenges and new opportunities in glioma chemotherapy
.
Front Oncol
.
2019
;
9
:
1547
.
62.
Murakami
M,
Mizutani
A,
Asano
S,
et al
A mechanism of acquiring temozolomide resistance during transformation of atypical prolactinoma into prolactin-producing pituitary carcinoma: case report
.
Neurosurgery
.
2011
;
68
:
E1761
1767
;
discussion E1767.
63.
Hirohata
T,
Asano
K,
Ogawa
Y,
et al
DNA mismatch repair protein (MSH6) correlated with the responses of atypical pituitary adenomas and pituitary carcinomas to temozolomide: the national cooperative study by the Japan Society for Hypothalamic and Pituitary Tumors
.
J Clin Endocrinol Metab
.
2013
;
98
:
1130
1136
.
64.
Kamiya-Matsuoka
C,
Cachia
D,
Waguespack
SG,
et al
Radiotherapy with concurrent temozolomide for the management of extraneural metastases in pituitary carcinoma
.
Pituitary
.
2016
;
19
:
415
421
.
65.
Strosberg
JR,
Fine
RL,
Choi
J,
et al
First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas
.
Cancer
.
2011
;
117
:
268
275
.
66.
Kunz
PL,
Graham
N,
Catalano
PJ,
et al
A randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors: final analysis of efficacy and evaluation of MGMT (ECOG-ACRIN E2211)
.
J Clin Oncol
.
2022
;
40
:
4004
4004
.
67.
Zacharia
BE,
Gulati
AP,
Bruce
JN,
et al
High response rates and prolonged survival in patients with corticotroph pituitary tumors and refractory Cushing disease from capecitabine and temozolomide (CAPTEM): a case series
.
Neurosurgery
.
2014
;
74
:
E447
455
;
discussion E455.
68.
Thearle
MS,
Freda
PU,
Bruce
JN,
et al
Temozolomide (Temodar®) and capecitabine (Xeloda) treatment of an aggressive corticotroph pituitary tumor
.
Pituitary
.
2011
;
14
:
418
424
.
69.
Nakano-Tateno
T,
Satou
M,
Inoshita
N,
et al
Effects of CAPTEM (capecitabine and temozolomide) on a corticotroph carcinoma and an aggressive corticotroph tumor
.
Endocr Pathol
.
2021
;
32
:
418
426
.
70.
Cornell
RF,
Kelly
DF,
Bordo
G,
et al
Chemotherapy-induced regression of an adrenocorticotropin-secreting pituitary carcinoma accompanied by secondary adrenal insufficiency
.
Case Rep Endocrinol
.
2013
;
2013
:
675298
.
71.
He
L,
Forbes
JA,
Carr
K,
et al
Response of silent corticotroph pituitary carcinoma to chemotherapy: case report
.
J Neurosurg Sci
.
2016
;
60
:
272
280
.
72.
Hurel
SJ,
Harris
PE,
McNicol
AM,
et al
Metastatic prolactinoma: effect of octreotide, cabergoline, carboplatin and etoposide; immunocytochemical analysis of proto-oncogene expression
.
J Clin Endocrinol Metab
.
1997
;
82
:
2962
2965
.
73.
AbdelBaki
MS,
Waguespack
SG,
Salceda
V,
et al
Significant response of pituitary carcinoma to carboplatin, leucovorin and fluorouracil chemotherapy: a pediatric case report and review of the literature
.
J Neurooncol
.
2017
;
135
:
213
215
.
74.
Angelousi
A,
Dimitriadis
GK,
Zografos
G,
et al
Molecular targeted therapies in adrenal, pituitary and parathyroid malignancies
.
Endocr Relat Cancer
.
2017
;
24
:
R239
R259
.
75.
Lloyd
RV,
Scheithauer
BW,
Kuroki
T,
et al
Vascular Endothelial Growth Factor (VEGF) Expression in Human Pituitary Adenomas and Carcinomas
.
Endocr Pathol
.
1999
;
10
:
229
235
.
76.
Tanase
C,
Codrici
E,
Popescu
ID,
et al
Angiogenic markers: molecular targets for personalized medicine in pituitary adenoma
.
Per Med
.
2013
;
10
:
539
548
.
77.
Dai
C,
Liang
S,
Sun
B,
et al
Anti-VEGF therapy in refractory pituitary adenomas and pituitary carcinomas: a review
.
Front Oncol
.
2021
;
11
:
773905
.
78.
Ortiz
LD,
Syro
LV,
Scheithauer
BW,
et al
Anti-VEGF therapy in pituitary carcinoma
.
Pituitary
.
2012
;
15
:
445
449
.
79.
Touma
W,
Hoostal
S,
Peterson
RA,
et al
Successful treatment of pituitary carcinoma with concurrent radiation, temozolomide, and bevacizumab after resection
.
J Clin Neurosci
.
2017
;
41
:
75
77
.
80.
Lamb
LS,
Sim
HW,
McCormack
AI.
Case report: a case of pituitary carcinoma treated with sequential dual immunotherapy and vascular endothelial growth factor inhibition therapy
.
Front Endocrinol (Lausanne)
.
2020
;
11
:
576027
.
81.
Cooper
O,
Bonert
V,
Liu
NA,
Mamelak
AN.
Treatment of aggressive pituitary adenomas: a case-based narrative review
.
Front Endocrinol (Lausanne)
.
2021
;
12
:
725014
.
82.
Donovan
LE,
Arnal
AV,
Wang
SH,
Odia
Y.
Widely metastatic atypical pituitary adenoma with mTOR pathway STK11(F298L) mutation treated with everolimus therapy
.
CNS Oncol
.
2016
;
5
:
203
209
.
83.
Ilie
MD,
Lasolle
H,
Raverot
G.
Emerging, and novel treatments for pituitary tumors
.
J Clin Med
.
2019
;
8
:
E1107
.
84.
Marques
P,
Grossman
AB,
Korbonits
M.
The tumour microenvironment of pituitary neuroendocrine tumours
.
Front Neuroendocrinol
.
2020
;
58
:
100852
.
85.
Nie
D,
Xue
Y,
Fang
Q,
et al
Immune checkpoints: therapeutic targets for pituitary tumors
.
Dis Markers
.
2021
;
2021
:
5300381
.
86.
Mei
Y,
Bi
WL,
Greenwald
NF,
et al
Increased expression of programmed death ligand 1 (PD-L1) in human pituitary tumors
.
Oncotarget
.
2016
;
7
:
76565
76576
.
87.
Xi
Z,
Jones
PS,
Mikamoto
M,
et al
The upregulation of molecules related to tumor immune escape in human pituitary adenomas
.
Front Endocrinol (Lausanne)
.
2021
;
12
:
726448
.
88.
Nguyen
H,
Shah
K,
Waguespack
SG,
et al
Immune checkpoint inhibitor related hypophysitis: diagnostic criteria and recovery patterns
.
Endocr Relat Cancer
.
2021
;
28
:
419
431
.
89.
Dai
C,
Liang
S,
Sun
B,
Kang
J.
The progress of immunotherapy in refractory pituitary adenomas and pituitary carcinomas
.
Front Endocrinol (Lausanne)
.
2020
;
11
:
608422
.
90.
Majd
N,
Waguespack
SG,
Janku
F,
et al
Efficacy of pembrolizumab in patients with pituitary carcinoma: report of four cases from a phase II study
.
J Immunother Cancer
.
2020
;
8
:
e001532
.
91.
Naing
A,
Meric-Bernstam
F,
Stephen
B,
et al
Phase 2 study of pembrolizumab in patients with advanced rare cancers
.
J Immunother Cancer
.
2020
;
8
:
e000347
.
92.
Lin
AL,
Jonsson
P,
Tabar
V,
et al
Marked response of a hypermutated acth-secreting pituitary carcinoma to ipilimumab and nivolumab
.
J Clin Endocrinol Metab
.
2018
;
103
:
3925
3930
.
93.
Sol
B,
de Filette
JMK,
Awada
G,
et al
Immune checkpoint inhibitor therapy for ACTH-secreting pituitary carcinoma: a new emerging treatment?
Eur J Endocrinol
.
2021
;
184
:
K1
K5
.
94.
Duhamel
C,
Ilie
MD,
Salle
H,
et al
Immunotherapy in corticotroph and lactotroph aggressive tumors and carcinomas: two case reports and a review of the literature
.
JPM
.
2020
;
10
:
88
.
95.
Goichot
B,
Taquet
MC,
Baltzinger
P,
et al
Should pituitary carcinoma be treated using a NET-like approach? A case of complete remission of a metastatic malignant prolactinoma with multimodal therapy including immunotherapy. Clin Endocrinol (Oxf). Published online Nov 29,
2021
.
96.
Ilie
MD,
Vasiljevic
A,
Jouanneau
E,
Raverot
G.
Immunotherapy in aggressive pituitary tumors and carcinomas: a systematic review
.
Endocr Relat Cancer
.
2022
;
29
:
415
426
.
97.
Yeung
JT,
Vesely
MD,
Miyagishima
DF.
In silico analysis of the immunological landscape of pituitary adenomas
.
J Neurooncol
.
2020
;
147
:
595
598
.
98.
Bai
R,
Lv
Z,
Xu
D,
Cui
J.
Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitors
.
Biomark Res
.
2020
;
8
:
34
.
99.
Shao
C,
Li
G,
Huang
L,
et al
Prevalence of high tumor mutational burden and association with survival in patients with less common solid tumors
.
JAMA Netw Open
.
2020
;
3
:
e2025109
e2025109
.
100.
Jardim
DL,
Goodman
A,
de Melo Gagliato
D,
Kurzrock
R.
The challenges of tumor mutational burden as an immunotherapy biomarker
.
Cancer Cell
.
2021
;
39
:
154
173
.
101.
Yu
Y,
Villanueva-Meyer
J,
Grimmer
MR,
et al
Temozolomide-induced hypermutation is associated with distant recurrence and reduced survival after high-grade transformation of low-grade IDH-mutant gliomas
.
Neuro Oncol
.
2021
;
23
:
1872
1884
.
102.
McGrail
DJ,
Pilié
PG,
Rashid
NU,
et al
High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types
.
Ann Oncol
.
2021
;
32
:
661
672
.
103.
Ilie
MD,
Villa
C,
Cuny
T,
et al
Real-life efficacy, and predictors of response to immunotherapy in pituitary tumors: a cohort study. Eur J Endocrinol. Published online Septr
2022
.
104.
Gatto
F,
Arvigo
M,
Ferone
D.
Somatostatin receptor expression and patients' response to targeted medical treatment in pituitary tumors: evidences and controversies
.
J Endocrinol Invest
.
2020
;
43
:
1543
1553
.
105.
Bodei
L,
Kidd
M,
Paganelli
G,
et al
Long-term tolerability of PRRT in 807 patients with neuroendocrine tumours: the value and limitations of clinical factors
.
Eur J Nucl Med Mol Imaging
.
2015
;
42
:
5
19
.
106.
Giuffrida
G,
Ferraù
F,
Laudicella
R,
et al
Peptide receptor radionuclide therapy for aggressive pituitary tumors: a monocentric experience
.
Endocr Connect
.
2019
;
8
:
528
535
.
107.
Novruzov
F,
Aliyev
JA,
Jaunmuktane
Z,
et al
The use of 68Ga DOTATATE PET/CT for diagnostic assessment and monitoring of 177Lu DOTATATE therapy in pituitary carcinoma
.
Clin Nucl Med
.
2015
;
40
:
47
49
.
108.
Maclean
J,
Aldridge
M,
Bomanji
J,
et al
Peptide receptor radionuclide therapy for aggressive atypical pituitary adenoma/carcinoma: variable clinical response in preliminary evaluation
.
Pituitary
.
2014
;
17
:
530
538
.
109.
Lin
AL,
Tabar
V,
Young
RJ,
et al
Synergism of checkpoint inhibitors and peptide receptor radionuclide therapy in the treatment of pituitary carcinoma. J Endocr Soc.
2021
;
5:bvab133.
110.
Hou
J,
Yang
Y,
Chen
N,
et al
Prognostic value of volume-based parameters measured by SSTR PET/CT in neuroendocrine tumors: a systematic review and meta-analysis
.
Front Med (Lausanne)
.
2021
;
8
:
771912
.
111.
Kratochwil
C,
Stefanova
M,
Mavriopoulou
E,
et al
SUV of [68Ga]DOTATOC-PET/CT predicts response probability of PRRT in neuroendocrine tumors
.
Mol Imaging Biol
.
2015
;
17
:
313
318
.
112.
Alevroudis
E,
Spei
ME,
Chatziioannou
SN,
et al
Clinical utility of 18F-FDG PET in neuroendocrine tumors prior to peptide receptor radionuclide therapy: a systematic review and meta-analysis
.
Cancers (Basel)
.
2021
;
13
:
1813
.
113.
Cremonesi
M,
Ferrari
ME,
Bodei
L,
et al
Correlation of dose with toxicity and tumour response to 90Y- and 177Lu-PRRT provides the basis for optimization through individualized treatment planning
.
Eur J Nucl Med Mol Imaging
.
2018
;
45
:
2426
2441
.

Source of Support: None. Conflict of Interest: None.

This work is published under a CC-BY-NC-ND 4.0 International License.