Amsterdam Medical Student journal (AMSj) | Volume 10

Page 1

Articles | cHANGING PERSPECTIVES | Subject 101 | Trial and error | SOLVING STATISTICS interview | clinical image | radiology image | EXPERT OPINION

Amsterdam Medical Student journal

EDITION 10 | FEBRUARY 2018


1

Index 2

Editorial

3

Selective Serotonine Reuptake Inhibitors during Pregnancy and Persistent Pulmonary Hypertension of the newborn | S. Bredenbeek, S.D. Sie

9

Clinical image | A child with kidney stones

10

Changing perspectives | The MRI and arthroscopy of the knee in patients over 50 years of age

11

Radiology image | A woman with dysmenorrhea and hypermenorrhea

12

Targeted temperature Management after cardiac arrest: 33˚C versus 36˚C hypothermia after an out-of-hospital cardiac arrest | B. ten Haaft, P. Thoral

17

Solving statistics | Help! How do I get a normal distribution?

19

Interview | Prof. dr. U. Beuers

21

Subject 101 | Glucosuria – trick or treatment?

23

Adoptive cell therapy using tumor infiltrating lymphocytes versus marrow infiltrating lymphocytes in the treatment of cancer | M. de Jong

28

Hyperthermic intraperitoneal chemotherapy for patients with peritoneal metastases of colorectal cancer treated with adjuvant chemotherapy | Y. Salhi

37

Expert opinion | Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease

38

Answers | Clinical image

39

Thinking about tomorrow: A literature review on the long-term impact of adolescent cannabis use on cognition and intelligence | T.E.K. van Os, T.J. de Vries

45

Answers | Radiology image

AMSj Vol. 10 | February 2018


2

Editorial Since 1901 the Nobel Prize praises excellent researchers for their contributions in various disciplines in the (bio)medical field. Until now, three Dutch scientists received the Nobel Prize in Physiology or Medicine. Willem Einthoven, who discovered the mechanism of the electrocardiogram, was the first of them to receive the golden medal and prize money to the value of what would now be about 1 million euros in 1924. Shortly after, Christiaan Eijkman received the award in 1924 (together with Frederick Gowland Hopkins, Great Britain) for his discovery in the role of vitamins in Beriberi. The last Dutch researcher who received the Nobel Prize in Physiology or Medicine was Niko Tinbergen, who received the award together with Karl von Frisch and Konrad Lorenz (both from Austria) in 1973 for their analysis of social behaviour in animals. Over the years researchers worked tremendously hard to understand our world a little better. Sometimes things are discovered by accident, such as the discovery of the world’s first antibiotic, by Alexander Fleming. In either case, multiple steps are required to eventually find the last piece for ground-breaking research. AMSj is lucky to contribute to this process and we are happy to present our 10th edition, in which varied topics of research are covered. S. Bredenbeek and S.D. Sie review the risk of persistent pulmonary hypertension of the newborn after exposure to SSRIs during pregnancy. B. ten Haaft and P. Thoral assess neurologic functions in patients with out-of-hospital cardiac arrests after different targeted temperature managements.

Y. Salhi and colleagues assess the benefits of adjuvant chemotherapy in patients with isolated peritoneal carcinomatosis of colorectal cancer treated with hyperthermic intraperitoneal chemotherapy (HIPEC). Finally, T.E.K. van Os and T.J. de Vries accentuate the long-term effects of cannabis use by adolescents in cognitive functioning and intelligence. In 2018 AMSj will continue publishing your research, albeit with a brand new lay-out! What will 2018 bring you? Consider publishing your research in AMSj and you might get one step closer to what might be a Nobel Prize..! Vera de Jonge Student Editor in Chief (VUmc)

Over the years cancer has been a topic of growing interest in the research field of medicine. In 2018, we can expect an ongoing accumulation in cancer knowledge. In this AMSj edition M. de Jong analyses adoptive cell therapy, a powerful method of immunotherapy, in patients with cancer. What is the difference between adoptive cell therapy using tumor-infiltrating lymphocytes and adoptive cell therapy using marrow-infiltrating lymphocytes? With colorectal cancer being one of the most common cancers in the modern western world, February 2018 | AMSj Vol. 10


3 ARTICLE

Selective Serotonin Reuptake Inhibitors during pregnancy and Persistent Pulmonary Hypertension of the Newborn: a systematic review S. Bredenbeek, S.D. Sie Department of Neonatology, VU University Medical Center, Amsterdam

ABSTRACT

B a c kg r o u n d Depression during pregnancy is common and mostly treated with Selective Serotonin Reuptake Inhibitors (SSRIs). It is well known that exposure to SSRIs during pregnancy can result in Poor Neonatal Adapation (PNA). In addition, animal studies suggest that intrauterine exposure to SSRIs is associated with Persistent Pulmonary Hypertension of the Newborn (PPHN), perhaps because serotonin affects the pulmonary vasculature. PPHN is a severe disorder leading to severe hypoxemia in all organs of the newborn and has a high mortality rate. A i m The aim of this review is to analyze the association between exposure to SSRIs during pregnancy and the occurrence of PPHN. Furthermore, we analyzed the occurrence of PPHN after SSRI use during early versus late pregnancy and between the specific SSRIs. M e t h o d s Pubmed was searched for original researches concerning exposure to SSRIs during pregnancy and PPHN. R e s u lt s Nine studies were included in this review. Most studies were retrospective cohort or case control studies. Seven studies conclude there is an increased risk for PPHN after the use of SSRIs during pregnancy, especially during late pregnancy. Two studies showed no relationship between exposure to SSRIs during pregnancy and PPHN. However, these two studies did not adjust for potential confounders or had a small number of participants. The influence of a specific type of SSRI on PPHN was analyzed in two studies. Both concluded that exposure to paroxetine, followed by sertraline, resulted in an increased risk for PPHN compared to other SSRIs. C o n c l u s i o n Overall, most studies found an increased risk for PPHN after exposure to SSRIs during late pregnancy. Although the absolute risk is small, the risk for PPHN after the use of SSRIs during pregnancy should be taken into account and precautions should be taken.

INTRODUCTION

Depression during pregnancy is common and occurs in 7% to 15% of all pregnant women and approximately 3% to 13% is treated with antidepressants, mostly Selective Serotonin Reuptake Inhibitors (SSRIs).1,2 Also, SSRIs are prescribed for other disorders, like anxiety or obsessive-compulsive disorders. After prenatal exposure to SSRIs, infants are at risk to develop symptoms of Poor Neonatal Adaptation (PNA).3 The etiology of PNA is still unknown and the most common symptoms are feeding and sleeping difficulties, tremors and respiratory distress.4 Another possible effect of in utero exposure to SSRIs is the occurrence of Pulmonary

Persistent Hypertension of the Newborn (PPHN), as serotonin is a potent pulmonary vasoconstrictor. Recently, an animal study demonstrated a major ductus arteriosus (DA) constriction in utero in mice, prenatally exposed to SSRIs.5 Another study showed that SSRIs increased fetal pulmonary vascular resistance in sheep.6 These findings suggest that SSRIs influence pulmonary vasculature and therefore can contribute to PPHN. PPHN is a severe disorder of the pulmonary vasculature, in which relaxation in the fetal pulmonary arteries fails after birth, causing elevated pressure in the pulmonary arteries.7,8 This leads to right-to-left shunting via the ductus arteriosus or the foramen ovale, resulting in severe systemic hypoxemia.9

AMSj Vol. 10 | February 2018


4 ARTICLE

PPHN typically occurs in newborns with a gestational age of ≥34 weeks. PPHN has several causes, like infection or chronic intrauterine fetal hypoxia.7 Previous studies identified several maternal and fetal risk factors for PPHN, such as smoking and gender, and are therefore potential confounder.10,11 The prevalence of PPHN has been estimated at 1.9 per 1000 live births and has a high mortality from 10% to 60%, depending on the underlying disorder.9,12 Infants who survive are at risk for long-term effects, like chronic lung disease or neurodevelopmental problems.12-15 Because animal studies showed an effect of prenatal exposure to SSRIs on the pulmonary vasculature in the newborn, the aim of this literature review is to analyze the relationship between SSRIs and PPHN in humans. Furthermore, we analyzed the occurrence of PPHN after SSRI use during early versus late preganncy and between the specfic SSRIs.

METHODS

Pubmed was searched, using the keywords and Mesh-terms described in Table 1. Studies had to meet the following criteria: 1) The use of SSRIs during pregnancy 2) Original research 3) Published in English A first selection was made based on titles and abstracts. After reading the full text to assess eligibility of the selected studies, a second selection was made. Cross-references were checked to complete the selection.

RESULTS

The results of the search strategy are shown in Figure 1. In total, 9 studies were eligible from 76 studies found with the PubMed search. An overview of these studies can be found in appendix A1 and A2. Relation between the use of SSRIs regardless of the timing of exposure and PPHN

Three studies analyzed the relation between exposure to SSRIs regardless of the timing of exposure during pregnancy and PPHN. In a retrospective cohort study of Nörby et al.,17 736

newborns exposed to SSRIs and 723 304 non-exposed newborns were included. In the SSRI exposed group, 0.5% was diagnosed with PPHN, compared to 0.3% of the non-exposed infants. In this study, they adjusted for several factors, as described in Table 4. After adjustment, a significantly higher risk was found for PPHN after exposure to SSRIs during pregnancy (Odds Ratio (OR): 1.3 [95% Confidence Interval (CI): 1.0-1.6], p = 0.03).16 In another retrospective cohort study of Reis et al., an increased risk for PPHN after exposure to SSRIs was found (RR: 3.44 [95% CI: 1.49-6.79]). They included 14 979 infants exposed to SSRIs during pregnancy compared to 1 236 053 nonexposed infants.17 In contrast to the previous two studies, Chambers et al. did not find a relation between the use of SSRIs during pregnancy and PPHN. In total, 377 infants diagnosed with PPHN were included and compared to 836 controls. Among the infants with PPHN 4.2% was exposed to SSRIs compared to 2.9% of the controls (adjusted OR: 1.6 [95% CI: 0.8-3.2] and p = 0.16). They adjusted for several maternal factors (Table 4).18 Relation between the use of SSRIs during early and/or late pregnancy and PPHN

In total, all 9 included studies examined if the timing of exposure to SSRIs influences the occurrence of PPHN. All studies defined early pregnancy before the 20th week of gestation and late pregnancy after the 20th week of gestation. Nörby et al. found that the prevalence of PPHN was significantly higher after exposure to SSRIs during late pregnancy compared to early pregnancy (adjusted OR: 2.6 [95% CI: 1.4-4.8]).16 Bérard et al. included 143 281 pregnancies, of which 2184 were exposed during early pregnancy and 2184 exposed during late pregnancy. These groups were compared to non-exposed infants. In total, 0.5% of the exposed and 0.2% of the nonexposed infants were diagnosed with PPHN. A significantly higher risk for PPHN was verifiable after exposure to SSRIs during late pregnancy (adjusted OR: 4.29 [95% CI: 1.3413.77]). The use of SSRI during early pregnancy was not significantly associated with the risk of PPHN (adjusted OR: 0,37 [95% CI: 0.11- 1.22]).19 Huybrechts et al. included a total of 3 798 330

February 2018 | AMSj Vol. 10


5 ARTICLE Number of studies resulting from the search n=76

Excluded: n=67 - reviews (n=36) - did not concern PPHN (n=17) - comments (n=7) - animal studies (n=3) - no scientific articles (n=3) - did not concern SSRI (n=1)

Checking cross-references: No new or useful studies

Final selection n=9

FIGURE 1 Results of the search strategy

infants of whom 102 79 were exposed to SSRIs during late pregnancy. About 33.8 per 10 000 ([95% CI: 29.7-38.6]) infants exposed during late pregnancy compared to 24.9 per 10 000 ([95% CI: 23.7-26.1]) nonexposed infants were diagnosed with PPHN. The authors conclude that there is an association between the use of SSRIs during late pregnancy and PPHN (OR: 1.28 [95% CI: 1.01-1.64]). This OR was calculated after adjustment for primary PPHN.20 Kieler et al. compared 17 053 newborns exposed during early pregnancy only and 11 014 newborns exposed during late pregnancy. A significant higher risk for PPHN was ascertained after exposure to SSRIs during late pregnancy (OR: 2,1 [95% CI: 1.5-3.0]). This was also the case for exposure in early pregnancy (adjusted OR: 1,4 [95% CI: 1.0-2.0]), but more modest.21 Also Reis et al. found a significantly increased risk for PPHN after exposure to SSRIs during late (RR: 2.56 [95% CI: 1.17-4.85]) and early (RR: 2.30 [95% CI: 1.29-3.80]) pregnancy. A total of 10 170 infants exposed during early and 4809 infants exposed during late pregnancy are included.17 An increased risk for PPHN after exposure during early (n=7587) and late (n=2414) pregnancy was also found in Källén et al. After restriction to ≥34 weeks of gestation, there was a significant

increased risk for PPHN after exposure to SSRIs during early (RR: 2.38 [95% CI: 1.19-4.25]) and late (RR: 3.57 [95% CI: 1.16-8.33]) pregnancy. This increased risk remained significant after restriction to ≥37 weeks of gestation, respectively RR: 2.36 [95% CI: 1.08-4.78] and RR: 3.70 [95% CI: 1.01-9.48].22 The data of Chambers et al. found an association between exposure to SSRIs late in pregnancy and PPHN. In the group of neonates with PPHN, 16 neonates with PPHN were exposed to SSRIs. Of the 16 exposed infants diagnosed with PPHN, 12.5% (adjusted OR: 0.3 [95% CI: 0.1-1.2] and p= 0.08) were exposed during early pregnancy (adjusted OR: 0.3 [95% CI: 0.1-1.2] and p= 0.08) and 87.5% during late pregnancy (adjusted OR: 6.1 [95% CI: 2.2-16.8] and p = 0.001).18 On the contrary, no relationship between exposure to SSRIs during pregnancy and PPHN was found in the study of Wilson et al. and Andrade et al.23,24 None of the 20 infants diagnosed with PPHN in the study of Wilson et al. were exposed to SSRIs during late pregnancy (adjusted OR: 0 [95% CI: 0.0-3.0]).24 In the other study, 1104 exposed and 1104 non-exposed infants were analyzed. Of the infants exposed to SSRIs during the third trimester, 2.14 per 1000 ([95% CI: 0.26, 7.74]) were diagnosed with PPHN compared to 2.72 per 1000 ([95% CI: 0.56-7.93]) infants who were not exposed. The prevalence ratio for the association between PPHN and the use of SSRIs is 0.79 (95% CI: 0.07-6.89). Unlike almost all other previous studies, the results were not adjusted for possible confounders.23 Effects of individual SSRIs and dosage on occurrence of PPHN

Two studies mentioned the effect of individual SSRIs: Chambers et al. and Kieler et al.18,21 In the research performed by Chambers et al. the SSRIs paroxetine, sertraline, fluoxetine and citalopram were used. Only paroxetine, sertraline and fluoxetine were used after 20 weeks GA. The results of exposure to fluoxetine, sertraline and paroxetine during late pregnancy are shown in Table 2. Approximately 42.9% (3/7) infants exposed to fluoxetine, 77.8% (7/9) infants exposed to sertraline and 100% (4/4) exposed to paroxetine are diagnosed with PPHN in this

AMSj Vol. 10 | February 2018


6 ARTICLE Infants with PPHN (n=377)

Infants without PPHN (n=836)

% Diagnosed with PPHN after exposure

Fluoxetine

3

4

42.9%

Sertraline

7

2

77.8%

Paroxetine

4

0

100%

Table 2 Effects of fluoxetine, sertraline and paroxetine on the occurrence of PPHN (18)

Prevalence early pregnancy

Adjusted* OR OR (95% CI)

Prevalence late pregnancy

Adjusted* OR (95% CI)

Fluoxetine

1.8 per 1000

1.3 (0.6-2.8)

2.7 per 1000

2.0 (1.2-4.3)

Sertraline

2.5 per 1000

1.8 (1.1-3.0)

3.3 per 1000

2.3 (1.2-4.1)

Paroxetine

1.7 per 1000

1.3 (0.5-3.5)

3.9 per 1000

2.8 (1.2-6.7)

Sertraline

2.7 per 1000

1.9 (1.0-3.6)

3.5 per 1000

2.3 (1.3-4.4)

Escitalopram

0.4 per 1000

0.3 (0.0-2.2)

1.8 per 1000

1.3 (0.2-9.5)

Table 3 Effects of fluoxetine, citalopram, paroxetine, sertraline and escitalopram on the occurrence of PPHN (21) *Adjusted for maternal age, dispensed non-steroidal anti-inflammatory drugs and anti diabetic drugs, pre-eclampsia, chronic diseases during pregnancy, country of birth, birth year, level of delivery hospital, and birth order.

Adjustments:

Chambers et al.

Bérard et al.

Källén et al.

Nörby et al.

Reis et al.

Kieler et al.

Huybrechts et al.

Andrade et al.

Wilson et al.

Year of birth

No

Yes

Yes

Yes

Yes

Yes

Yes

No

Yes

Gestational age

No

No

Yes

Yes

Yes

Yes

Yes

No

Maternal age

No

Yes

Yes

Yes

Yes

Yes

Yes

No

Yes

Maternal smoking

Yes

Yes

Yes

Yes

Yes

Yes

No

No

Yes

Maternal BMI

Yes

Yes

Yes

Yes

Yes

Yes

No

No

Yes

Yes

Yes

Yes

Yes

Yes

Yes

No

Yes

No

Yes

Yes

No

No

Parity NSAID use

Yes

Other medication use

Yes

Yes

Yes

No

Yes

Yes

No

No

Previous maternal comorbidities

Yes

Yes

No

No

No

Yes

Yes

No

Yes

Mode of delivery

No

No

No

Yes

No

Yes

No

No

Yes

Severity of maternal depression

No

No

No

No

No

No

Yes

No

No

Association SSRIs exposure during late pregnancy and PPHN

Yes

Yes

Yes

Yes

Yes

Yes

Yes

No

No

OR/RR (95% CI)

OR: 5.1 (1.913.3)

OR: 4.29 (1.3413.77)

RR: 2.91 (0.0946.78)

OR: 2.6 (1.4-4.8)

RR: 2.56 (1.174.85)

OR: 2.1 (1.5-3.0)

OR: 1.28 (1.011.64)

RR: 0.79 (0.076.89)

OR: 0 (0.0-3.0)

Table 4 Adjustments for confounders used in the studies February 2018 | AMSj Vol. 10


7 ARTICLE

research. Although the absolute numbers are low, these results conclude the highest risk for PPHN after exposure to paroxetine.18 The research of Kieler et al. considered paroxetine, sertraline, citalopram, fluoxetine, fluvoxamine and escitalopram. According to these results, shown in Table 3, the risk for PPHN after exposure to SSRIs to 1 of these 5 SSRIs is always higher when exposed during late pregnancy. The highest risk for PPHN is after exposure to paroxetine, followed by sertraline. The risk for PPHN after exposure to escitalopram was not statistically significant. The prevalence of PPHN of the unexposed infants was in all cases 1.2 per 1000 infants.21 None of the authors analyzed the relationship between the dose of SSRIs and the occurrence of PPHN.

DISCUSSION

Most studies found an association between the use of SSRIs during pregnancy and the occurrence of PPHN. The two studies that concluded that there was no association between the use of SSRIs during pregnancy and PPHN had low numbers of infants participating in their research. Wilson et al. and Andrade et al. respectively included 20 and 1104 infants diagnosed with PPHN or exposed to SSRIs.23,24 In the remaining 7 studies the number of included infants exposed to SSRIs or diagnosed with PPHN varies between 377 and 102 179 infants.24 The association between SSRIs during pregnancy and the occurrence of PPHN was strongest after exposure during late pregnancy. The highest risk for PPHN was found after maternal use of paroxetine, compared to the other SSRIs. None of the studies analyzed the association between the dosage of the SSRI and the occurrence of PPHN. In general, most researches collected data about the prescription of SSRIs from different databases in different countries. All of these studies considered women with a prescription for SSRIs at least a year before pregnancy as use of SSRIs during pregnancy. However, it is not certain that those women really used SSRIs during pregnancy and in which stage of pregnancy. The International Statistical Classification of

Diseases and Related Health Problems (ICD) is used as a diagnostic method for PPHN in every included study. Furthermore, the definitions for early and late pregnancy used in each study were consistent. Almost all studies made adjustments for other causes of PPHN, such as meconium aspiration and cardiac malformations. Only in the study of KällÊn et al. it is not described if adjustments were made for other possible causes of PPHN.22 The applied adjustments for confounders are shown in Table 4. Most of the researches did adjust for many potential confounders, except Andrade et al.23 For example, mothers who used SSRIs during pregnancy were older, had more comorbidities together with a higher BMI (>30) and smoked more often.16,19-21 All of this can be a confounder in determining the causality of the PPHN, for which is not corrected in the research of Andrade et al.23 Despite the number of confounders for which is adjusted in most of the included studies, still a number of important aspects that can influence the results were not taken into consideration. Only one study analyzed the severity of the maternal depression and adjusted for this.20 However, it is not known if there is an association between maternal depression itself and PPHN. Three studies adjusted for mode of delivery as confounders or effect modifiers. This control for mode of delivery is essential for interpretation of the data, since there might be an association between the occurrence of PPHN and a caesarian section.25 Although the outcome measures were sometimes different (OR and RR), there is still no visible pattern between the amount of confounders for which is adjusted in the research and the outcome measure (see Table 4). For example, Kieler et al. adjusted for all possible confounders and calculated a lower OR than Chambers et al., that only adjusted for half as much confounders than Kieler et al.18,21 PPHN in preterms is mostly caused by underdeveloped, immature lungs. Therefore, most studies excluded preterms and adjust for GA as confounder. The results do not show if this assumption is correct. In total four studies did not adjust

AMSj Vol. 10 | February 2018


8 ARTICLE

for GA; two studies found the highest risk for PPHN after exposure to SSRIs during pregnancy. Based on the current literature, the occurrence of PPHN is especially higher after late exposure to SSRIs. This might be because of a stronger effect of serotonin on the pulmonary vasculature during late pregnancy compared to early pregnancy. Furthermore, it is notable that in both the study by Chambers et al. as the study by Kieler et al., the risk of PPHN was highest after exposure to paroxetine, followed by sertraline.18,21 It is possible that each SSRI has a different effect on the pulmonary vasculature and therefore on the occurrence of PPHN. To obtain more information about these different effects, further research is necessary.

4.

CONCLUSION

10.

Current literature suggests an association between the use of SSRIs during pregnancy and PPHN. However, the absolute risk remains small. An increased risk for PPHN is especially found after the use of SSRIs during late pregnancy and after exposure to paroxetine and sertraline.

5.

6.

7. 8. 9.

11.

12.

RECOMMENDATIONS

Based on the increased risk for PPHN after intrauterine exposure to SSRIs, it is essential to take this increased risk into account when prescribing an SSRI to pregnant women or women with a desire to have children. It is important to consider the effect of the severity of the maternal depression compared to the use of SSRIs during pregnancy. Although the absolute risk for PPHN is small, precautions should be taken. Possible precautions are delivery in the hospital and observing the infants for potential symptoms of PPHN. Sintha Sie, Pediatrician-Neonatologist VUmc, supported this systematic review.

REFERENCES

2. 3.

14.

15. 16. 17.

ACKNOWLEDGEMENTS

1.

13.

Toh S, Mitchell AA, Louik C, et al. Antidepressant use during pregnancy and the risk of preterm delivery and fetal growth restriction. J Clin Psychopharmacol. 2009;29(6):555-60. Evans J, Heron J, Francomb H, et al. Cohort study of depressed mood during pregnancy and after childbirth. BMJ. 2001;323(7307):257-60. Kieviet N, Dolman KM, Honig A. The use of psychotropic medication during pregnancy: how about the newborn? Neuropsychiatr Dis Treat. 2013;9:1257-66.

18.

19. 20.

21.

Kieviet N, Hoppenbrouwers C, Dolman KM, et al. Risk factors for poor neonatal adaptation after exposure to antidepressants in utero. Acta Paediatr. 2015;104(4):38491. Hooper CW, Delaney C, Streeter T, et al. Selective serotonin reuptake inhibitor exposure constricts the mouse ductus arteriosus in utero. Am J Physiol Heart Circ Physiol. 2016;311(3):H572-81. Delaney C, Gien J, Roe G, et al. Serotonin contributes to high pulmonary vascular tone in a sheep model of persistent pulmonary hypertension of the newborn. Am J Physiol Lung Cell Mol Physiol. 2013;304(12):L894-901. Dakshinamurti S. Pathophysiologic mechanisms of persistent pulmonary hypertension of the newborn. Pediatr Pulmonol. 2005;39(6):492-503. Fox WW, Duara S. Persistent pulmonary hypertension in the neonate: diagnosis and management. J Pediatr. 1983;103(4):505-14. E G, U N. Persistent Pulmonary Hypertension of the Newborn MSD manual, 2015. Reece EA, Moya F, Yazigi R, et al. Persistent pulmonary hypertension: assessment of perinatal risk factors. Obstet Gynecol. 1987;70(5):696-700. Van Marter LJ, Leviton A, Allred EN, et al. Persistent pulmonary hypertension of the newborn and smoking and aspirin and nonsteroidal antiinflammatory drug consumption during pregnancy. Pediatrics. 1996;97(5):65863. Walsh-Sukys MC, Tyson JE, Wright LL, et al. Persistent pulmonary hypertension of the newborn in the era before nitric oxide: practice variation and outcomes. Pediatrics. 2000;105(1 Pt 1):14-20. Glass P, Wagner AE, Papero PH, et al. Neurodevelopmental status at age five years of neonates treated with extracorporeal membrane oxygenation. J Pediatr. 1995;127(3):447-57. Clark RH, Kueser TJ, Walker MW, et al. Low-dose nitric oxide therapy for persistent pulmonary hypertension of the newborn. Clinical Inhaled Nitric Oxide Research Group. N Engl J Med. 2000;342(7):469-74. Farrow KN, Fliman P, Steinhorn RH. The diseases treated with ECMO: focus on PPHN. Semin Perinatol. 2005;29(1):8-14. Nörby U, Forsberg L, Wide K, et al. Neonatal Morbidity After Maternal Use of Antidepressant Drugs During Pregnancy. Pediatrics. 2016;138(5). Reis M, Källén B. Delivery outcome after maternal use of antidepressant drugs in pregnancy: an update using Swedish data. Psychol Med. 2010;40(10):1723-33. Chambers CD, Hernandez-Diaz S, Van Marter LJ, et al. Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N Engl J Med. 2006;354(6):579-87. Bérard A, Sheehy O, Zhao JP, et al. SSRI and SNRI use during pregnancy and the risk of persistent pulmonary hypertension of the newborn. Br J Clin Pharmacol. 2016. Huybrechts KF, Bateman BT, Palmsten K, et al. Antidepressant use late in pregnancy and risk of persistent pulmonary hypertension of the newborn. JAMA. 2015;313(21):2142-51. Kieler H, Artama M, Engeland A, et al. Selective serotonin reuptake inhibitors during pregnancy and risk of persistent pulmonary hypertension in the newborn: popu-

February 2018 | AMSj Vol. 10


9 clinical image

22.

23.

24.

25.

lation based cohort study from the five Nordic countries. BMJ. 2012;344:d8012. KällÊn B, Olausson PO. Maternal use of selective serotonin re-uptake inhibitors and persistent pulmonary hypertension of the newborn. Pharmacoepidemiol Drug Saf. 2008;17(8):801-6. Andrade SE, McPhillips H, Loren D, et al. Antidepressant medication use and risk of persistent pulmonary hypertension of the newborn. Pharmacoepidemiol Drug Saf. 2009;18(3):246-52. Wilson KL, Zelig CM, Harvey JP, et al. Persistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitors. Am J Perinatol. 2011;28(1):19-24. Grigoriadis S, Vonderporten EH, Mamisashvili L, et al. Prenatal exposure to antidepressants and persistent pulmonary hypertension of the newborn: systematic review and meta-analysis. BMJ. 2014;348:f6932.

A child with kidney stones A. Zwagemaker, M.J.S. Oosterveld

Case

This Syrian boy, the second child of consanguineous parents, presented with kidney stones in his country of origin at the age of 5½ years. In the following years he developed end stage kidney disease. He started hemodialysis treatment at the age of 11 years. In 2012 he underwent a kidney transplant, which failed soon after surgery. Hemodialysis was resumed. The following year he lost his ability to walk. In 2015 the patient moved to the Netherlands. Upon arrival, he was found to be underdialysed, underfed and to suffer from severe bone disease. An abdominal X-ray was performed.

Question

What is the diagnosis? A. Genitourinary tuberculosis B. Nephrocalcinosis C. Nephrolithiasis D. Urolithiasis

?

Answer on page 38 AMSj Vol. 10 | February 2018


10 CHANGING PERSPECTIVES

MRI and arthroscopy of the knee in patients over 50 years of age S.W. Hoeffelman & Dr. M.U. Schafroth

In the Netherlands knee injuries occur approximately 300,000 times a year.1 Meniscal tears are among the most common knee injuries with an incidence of 2 per 1,000 patients a year.2 As a result arthroscopic meniscectomy is performed nearly 30,000 times a year in patients over 50 years of age.3 In over half of these patients magnetic resonance imaging (MRI) was performed prior to surgery. However, multiple randomized controlled trial observed no significant difference between knee function, pain relief and patient satisfaction when comparing arthroscopy to conservative treatment.4,5 In addition, the amount of knee arthroscopies where no therapeutic intervention was conducted during surgery ranges from 27 to 61%.1 Consequently, it is questioned whether these procedures benefit healthcare efficacy. Especially when taking into account that patients over 50 years of age are at risk of having existing osteoarthritis (OA) of the knee.6 This is accompanied by degenerative meniscal tears in up to 80%.7 Therefore, differentiating whether the symptoms are caused by the meniscal tear or developing OA is quite challenging. As a result, there is no indication to perform MRI in patients over 50 years of age according to the Netherlands Orthopedic Association (NOV). On that account, it is advised only to perform knee arthroscopy in case of locking of the knee caused by mechanical obstruction in these patients. This is without performing MRI prior to surgery.1

References

1. 2.

3.

4. 5.

6. 7.

NOV. Richtlijn Artroscopie van de knie, 2010. Van der Linden, M. W., Westert, G. P., de Bakker, D. H., & Schellevis, F. G. Tweede Nationale Studie naar ziekten en verrichtingen in de huisartspraktijk: klachten en aandoeningen in de bevolking en in de huisartspraktijk. Utrecht/Bilthoven: NIVEL/RIVM;2004. CBS. Operaties in het ziekenhuis; soort opname, leeftijd en geslacht, 1995-2010, retrieved via http://statline. cbs.nl/StatWeb/publication/?VW=T&DM=SLNL&PA=80386NED&LA=NL. Crevoisier, X., Munzinger, U., & Drobny, T. Arthroscopic partial meniscectomy in patients over 70 years of age. Arthroscopy, 17, 2001. p.732-736. Herrlin, S., Hallander, M., Wange, P., Weidenhielm, L., & Werner, S. Arthroscopic or conservative treatment of degenerative medial meniscal tears: a prospective randomised trial. Knee.Surg.Sports Traumatol.Arthrosc., 15, 2007. p. 393-401. Van Bodegom-Vos, L., Nelissen, R.G.H.H., Diercks, R.L. Project: Step-down knee MRI and ARThroscopy for orthopaedic patients > 50 years (SMART). Suter, L. G., Fraenkel, L., Losina, E., Katz, J. N., Gomoll, A. H., & Paltiel, A. D. Medical decision making in patients with knee pain, meniscal tear, and osteoarthritis. Arthritis Rheum., 61, 2009. p.1531-1538.

Therefore a substantial reduction of MRI and knee arthroscopies in patients over 50 years of age is warranted, especially in these times of increasing awareness of cost-effectiveness.6

February 2018 | AMSj Vol. 10


11 RADIOLOGY image

A woman with dysmenorrhea and hypermenorrhea S. Spijkers & M. Maas

Patient

• • • •

Age: 42 Gender: female Medical history: none Initial presentation: heavy, painful and elongated menstrual period for a few years.

Checklist MRI female pelvis

1. • • • • 2. • • 3. • 4. • • 5. • • •

Uterus Size and position Endometrium: thickness, homogeneity, focal lesions, intraluminal fluid, IUD Inner myometrium (junctional zone): thickness, sharpness of border, intramyometrial hyperintensities Caesarean section defect: consider the medical history Cervix Mucosa: thickness, cysts, luminal fluid Stroma: integrity, parametrial infiltration Vagina Cystic lesions, vaginal wall, foreign bodies Adnexa Ovaries: dimensions, cystic lesions, solid lesions Parovarian region: cystic lesions, vascular lesions, lymphadenopathy Other anatomy Bladder Bowel Musculoskeletal structures: bony pelvis, lower lumbar spine, muscles, tendons

Question 1: Which MRI sequences are shown? a) T1 weighted b) T2 weighted c) T1 fat sat weighted d) T2 fat sat weighted Question 2: Which anomaly is visible on the MRI image? a) Increased thickness of the inner myometrium (junctional zone) b) Focal lesions of the endometrium c) Wrongly placed IUD d) None Hint: Follow the checklist to assess the image of the uterus, consider the difference of the myometrium and the endometrium.

Question 3: What may underpin the hyperintensities visual in the uterus on the MRI image? a) Fat b) Debris c) Blood d) Artefacts Hint: consider the properties of the MRI image, the hyperintensities can be found in the myometrium. Question 4: What is the diagnosis? a) Endometriosis b) Adenomyosis c) Uterine fibroids d) Myoma Hint: Take into account the anatomical location of the anomalies. Question 5: What is the modality of choice to diagnose this condition? a) Vaginal echo b) Hysterosalpingogram c) CT d) MRI Hint: Take into account the anomalies you have seen. By which imaging technique would they have been visualised most optimally?

AMSj Vol. 10 | February 2018

?

Answer on page 45


12 ARTICLE

Targeted Temperature Management after cardiac arrest: 33˚C versus 36˚C hypothermia after an out-of-hospital cardiac arrest – a review B. ten Haaft, BSc, P. Thoral, MD Department of Intensive Care, VU University Medical Centre, Amsterdam, The Netherlands

Abstract

B a c kg r o u n d Survivors of out-of-hospital cardiac arrest have a high risk of death and have a high risk for poor neurologic function compared to people who never have had a cardiac arrest. Previous trials have shown beneficial results in patients who were treated with therapeutic hypothermia compared to standard treatment. Nevertheless, due to a lack of research, evidence for the target temperature associated with the best outcome is limited. The objective of this review is to discover the difference in neurologic function by the Cerebral Performance Categories (CPC) and mortality after targeted temperature management with 33̊C compared to 36̊C. M e t h o d s Five studies were identified by a computerized search on PubMed. All studies researched the different targeted temperatures and the corresponding neurologic outcome and mortality rate. Systematic reviews were excluded and unconscious patients with an out-of-hospital cardiac arrest older than 18 years old were included. R e s u lt s In total, five articles were included in this review. One article compared the neurologic function and mortality of targeted temperature management (TTM) at 33˚C and 36˚C, but concluded no difference in outcome between 33˚C and 36˚C. The other four articles examined a targeted temperature between 32˚C and 37˚C. All four articles supported therapeutic hypothermia at 33/34˚C. The CPC 1-2 and mortality of 33˚C did not show high discrepancy compared to 36˚C. C o n c l u s i o n The best neurologic outcome and mortality rate after targeted temperature management can neither be associated with 33˚C nor with 36˚C.

Introduction

Survivors of out-of-hospital cardiac arrest (OHCA) are thought to have high risks of death and poor neurologic function. Organ dysfunction is also related to out-of-hospital cardiac arrests.1 The overall incidence of out-of-hospital cardiac arrests in Europe equals 37.7 per 100,000 people per year with a survival of 10,7%.2 Since this survival rate is very low, new and effective procedures that may be applied after cardiac arrest (CA) are desired. Suggested by animal experiments, neuronal damage is reduced after treatment with hypothermia.3,4 In the same way, patients should not be exposed to hyperthermia after return of spontaneous circulation (ROSC) in OHCA since hyperthermia leads to unfavourable neurologic outcomes.5 Death could be prevented by a successful period of resuscitation. However, most deaths occur after resuscitation due to the development of irreversible neurological

dysfunction during the no-flow and ROSC period. In these periods, free oxygen radicals and other mediators are generated and released into the blood flow which could lead to damage of the brain and surrounding neurons.6 Bernard and colleagues (2002) show a beneficial neurologic result in patients treated 12 to 24 hours with targeted temperature management (TTM) compared to standard treatment.14 Despite the fact that most physiologic evidence advocates TTM, there is not enough knowledge regarding the optimal targeted temperature. The American Heart Association guideline advises 33̊C as the optimal targeted temperature after an out-of-hospital cardiac arrest.7 However, current guidelines used in Dutch hospitals present no explicit temperature preference, which means hospitals form their own standards regarding targeted temperature management.8 In addition, due

February 2018 | AMSj Vol. 10


13 ARTICLE

to a lack of research, the target temperature associated with the best outcome remains unknown. The aim of this review is to assess the effect of targeted temperature management at 33˚C versus 36˚C on neurologic outcome according to Cerebral Performance Category (CPC)9 and mortality in patients after an out-of-hospital cardiac arrest. We hypothesized that targeted temperature management with a targeted temperature of 33̊C has better neurological outcome compared to a target temperature of 36̊C since we expect lower body temperatures to be associated with a lower energy requirement and therefore less ischemia and less neurologic dysfunction.

Methods

Search Strategy

PubMed was searched for articles published from January 2000 until June 2017. The search was performed using the following keywords, broadened by its MeSH terms: “Out of hospital cardiac arrest“ and “Targeted Temperature Management”. The final search details are shown in the supplemental data. For the selection of eligible studies, articles must contain information about out-of-hospital cardiac arrests and hypothermia. Moreover, articles were assessed by inclusion and exclusion criteria such as age, subject group and follow-up time. An additional search was done according to the snowball strategy. The Nielsen, et al. (2013)10 TTM trial, Esteban Lopez-de- Sa, et al. (2012)11, Jin Joo Kim, et al. (2011)12, Michael Holzer, et al. (2002)13 and Stephen Bernard, et al. (2002)14, used several appropriate references for this review.The primary outcome of interest was favourable neurologic function. The Cerebral Performance Category was used to define neurologic outcome and ranges from 1 to 5, with 1 representing a good cerebral performance, 2 moderate disability, 3 severe disability, 4 coma and 5 brain death (Table 1).15 In this review, the CPC was divided into two scales: CPC 1-2 that refers to a good and favourable outcome and CPC 3-5 that refers to a poor and unfavourable outcome. Since in some cases the CPC was not divided into two categories, calculations were done to make this partition possible. CPC 1-2 and CPC 3-5 are expressed in percentage as a comparison to the whole group of

Cerebral performance categories 1. Good Cerebral Performance (normal life)

Conscious, alert

2. Moderate Cerebral Disability

Conscious, disabled but independent

3. Severe Cerebral Disability

Conscious, disabled and dependent

4. Coma/Vegetative State

Unconscious, unaware and no cognition

5. Brain Death

Certified brain dead or dead by traditional criteria

Table 1 Cerebral performance categories.

subjects per study. The secondary outcome was mortality expressed in percentage. Inclusion and Exclusion Criteria

Articles were included if the subjects were older than 18 years old and consisted of unconscious/ comatose patients. Most importantly, studies were included when the cardiac arrest had taken place outside the hospital (OHCA) and neurologic function and mortality had been measured after a minimum of 30 days follow-up. Systematic reviews were excluded, as well as articles published before the year 2000, since the only the most recent information should be included.

Results

Characteristics of Included Trials

In total, 81 hits were found, of which five articles met the inclusion criteria. Results of these five studies are shown in Table 2 to Table 5. Some values were not found in the text; we made calculations in order to include these results in our tables. A recent TTM trial, by Nielsen et al. (2013), examined targeted temperature management at 33˚C and 36˚C. The four other studies, Esteban Lopez-de- Sa, et al. (2012), Jin Joo Kim, et al. (2011), Michael Holzer, et al. (2002) and Stephen Bernard, et al. (2002), focused on hypothermia after a cardiac arrest but did not explicitly study 33˚C and 36˚C. Other temperatures with a range from 32˚C to 37+˚C were also examined and used in this review to create an overall view of the different targeted temperatures and their neurologic function as a result. Most studies compared two targeted temperatures10-12, 14 and only one

AMSj Vol. 10 | February 2018


14 ARTICLE Authors (year)

Study populations

Methods

Time to BLS/ ALS/ROS

Main results related to the research question

Niklas Nielsen, MD, Ph.D, et al. (2013)

N=939, 473pt 33˚C group, 466 pt 36˚C group, unconscious (<8 GCS), 18+ years old.

RCT, 33˚C vs. 36˚C, 180 days follow-up

33: 1/10/25 36: 1/9/25

TTM at a targeted temperature of 33˚C is not beneficial compared to a targeted temperature of 36˚C.

Esteban Lopez-deSa, MD, FESC, et al. (2012)

N=36, 18 pt 32˚C group, 18 pt 34˚C group, 18+ years old.

RCT, 32˚C vs. 34˚C, 180 days follow-up

32: 13/9,6/21 34: 12/9,8/32

A lower targeted temperature might be associated with a better outcome.

Jin Joo Kim, MD, et al. (2011)

N=62, 44 male and 18 female. 13 pt group 32˚C, 21 pt group 33˚C, 28 pt group 34˚C

Prospective study, 32˚C vs. 33˚C vs. 34˚C, 30 days follow-up

32: -/14/24 33: -/14/34 34: -/14/34

33˚C and 34˚C is associated with better outcomes in patients after an OHCA than 32˚C.

Michael Holzer, MD, et al. (2002)

N=275, 137 pt 33˚C group, 138 pt normothermia group, 18+ years old

RCT, 33˚C vs. normothermia, 180 days follow-up

33: -/-/21 37: -/-/22

MRH is associated with positive neurologic outcomes and mortality.

Stephen Bernard, et al. (2002)

N=77, 43 pt 33˚C group 34 pt 37˚C group.

RCT, 33˚C vs. 37˚C, 1-30 days follow-up

33: -/10/27 37: -/11/25

TTM reduces neurologic damage in patients after OHCA.

Table 2 Clinical Characteristics of the 10 Included Articles.

study compared three targeted temperatures.13 All studies apply TTM for 12h to 24h but not all studies use the same method for reaching their target temperature. Ice packs, helmets and invasive treatment were used. Clinical characteristics are presented in Table 2. No distinction was made between the causes of cardiac arrest (CA). Time to return of spontaneous circulation (ROSC), time to basic life support (BLS) and time to advanced life support are presented in Table 2. Clinical Outcomes

All neurologic outcomes are presented in Table 3. Favourable neurologic outcomes reach from 7.7% to 66%. The TTM trial done by Niklas N. et al. (2013) shows no significant difference in neurologic outcome between therapeutic hypothermia at 33˚C and 36˚C (p=0.78), while the second study done by Lopez-de-Sa E. et al. (2012) presents better neurologic outcomes related to 33˚C, 50% against 22.2% at 36˚C. Considering Joo Kim J. et al. (2011), 34˚C shows better neurologic outcomes compared to 32˚C and 33˚C. Concerning Holzer M. et al. (2002), therapeutic hypothermia results in significant higher favour-

able neurologic outcomes (p=0.009) but also results in a significant higher mortality rate of 55% compared to 41% at 33˚C (p=0.02). Lastly, Bernard S. et al. (2002) reports better neurologic outcomes as well as lower mortality rates associated with TTM at 33˚C (respectively p=0.046 and p=0.145). Concerning mortality, no clinically significant differences between the studies were observed. Adverse events

Table 5 presents the most severe adverse events after hypothermia and cardiac arrests including (septic) shock, renal failure and arrhythmias. The Supplementary Appendix of Nielsen N, et al. (2013)16 shows no major differences in adverse events after hypothermia. Moreover, Bernard S, et al. (2002), suggests no significant differences in adverse events between 33˚C and 36˚C as well.

Discussion

In this review we analysed studies about neurologic function and mortality after TTM at 33 and 36 in patients after OHCA. There are no

February 2018 | AMSj Vol. 10


15 ARTICLE

Niklas N. 2013 Lopez-de-Sa E. 2012 Joo Kim J. 2011 Holzer M. 2002 Bernard S. 2002

Type of measurement

32˚C

33˚C

34˚C

35˚C

36˚C

37˚C+

CPC 1-2 (%)

-

46.0

-

-

48.0

-

CPC 3-5 (%)

-

54.0

-

-

52.0

-

CPC 1-2 (%)

50.0

-

22.2

-

-

-

CPC 3-5 (%)

50.0

-

77.8

-

-

-

CPC 1-2 (%)

7.7

19.0

32.1

-

-

-

CPC 3-5 (%)

92.3

81.0

67.9

-

-

-

CPC 1-2 (%)

-

55

-

-

39

CPC 3-5 (%)

-

45

-

-

61

CPC 1-2 (%)

-

49

-

-

-

26

CPC 3-5 (%)

-

51

-

-

-

74

P Value 0.78 0.08 0.196 0.009 0.046

Table 3 Neurologic Outcomes.

clinically significant differences noticed in neurologic function and mortality between the two targeted temperatures. There is no consistency in the main results of the studies; not every article led to the same conclusion. The main limitation of this study is the heterogeneity of articles in terms of clinical details, which may, for example, lead to an influence of age on neurologic outcomes known as effect modification. Moreover, every study implicates their own cooling technique since targeting at an exact temperature and maintaining this temperature within narrow limits is difficult. These results therefore need to be interpreted with caution. Different techniques for hypothermia might influence results as well as timing of TTM and duration of induced hypothermia that can differ from 12h to 24h. High discrepancy is observed regarding the cause of CA. Some studies exclude asystole10 and other studies 11-14 include non-shockable rhythms or only ventricular fibrillation. Ventricular fibrillation is known to be associated with a lower CPC compared to other rhythms such as asystole or pulseless electrical activity. Consequently, false favorable neurologic outcomes or lower mortalities could be registered. All these details may contribute to heterogeneous outcomes, but those factors also increase the generalizability of the findings.

The first 6 minutes after a cardiac arrest are crucial for neurons and brain cells.17 Moreover, the time interval from collapse to initiation of BLS is the most powerful determent of restoration of a beating heart. Other time intervals contribute to this as well such as time to ROSC.18 This suggests the importance of time in minutes till the start of basic life support. Different times to BLS were reported such as a mean of 1 minute according to Niklas N. et al. (2013). Unfortunately, not many studies were able to report results regarding this factor. Nevertheless BLS within 1 minute is a number with high ambition and besides often not in public obtainable. Notwithstanding the fact that therapeutic hypothermia certainly influences neurologic function, we must consider the impact of the different times to BLS since naturally a treatment may have a larger therapeutic impact, when the (neurologic) condition is more severe. Therefore, it is important to bear in mind the possible bias in these studies.

Conclusion

Our study is not in line with several major studies, which suggested that targeted temperature management at 33˚C has an effective part of treatment in post resuscitation care.10, 19-21 The best neurologic outcome and mortality rate after targeted temperature management can neither be associated with 33˚C nor with 36˚C. Future research programs should compare the neurologic function of patients after targeted temperature management at 33˚C and 36˚C and pay attention to the time to

AMSj Vol. 10 | February 2018


16 ARTICLE

Niklas N. 2013

32˚C

33˚C

34˚C

35˚C

36˚C

37˚C

P Value

-

50.0

-

-

48.0

-

0.51

Lopez-de-Sa E. 2012

55.6

-

88.9

-

-

-

0.03

Joo Kim J. 2011

23.1

52.4

35.7

-

-

-

0.212

Holzer M. 2002

-

41.0

-

-

-

55.0

0.02

Bernard S. 2002

-

51

-

-

-

68

0.145

Table 4 Mortality (%). Body temperature (˚C) Niklas N. 2013 Lopez-de-Sa E. 2012 Joo Kim J. 2011

Holzer M. 2002 Bernard S. 2002

Renal Replacement Therapy (%)

(Septic) Shock (%)

Arrythmia (%)

33

11

4.8

38.1

36

9.1

5.4

31.1

32

11

N.A.

77.8

34

0

N.A.

22.2

32

15.4

15.4

15.4

33

19.0

4.76

23.8

34

7.14

3.57

17.9

33

13

10

36

37

7

10

32

33

0

0

N.A.

37

0

0.74

N.A.

TABLE 5 Adverse events. Abbreviations: N.A. = Not applicable

BLS, quality of life and cause of cardiac arrest. Targeted temperature management should focus on avoiding hyperpyrexia as main goal.

References

1.

2. 3.

4.

5.

Moulaert VR, Verbunt JA, van Heugten CM, et al. (2009). Cognitive impairments in survivors out-of-hospital cardiac arrest: a systematic review. Resuscitation; 80:297-305 Atwood C, Eisenberg MS, Herlitz J, et al. (2005). Incidence of EMS-treated out-of-hospital cardiac arrest in Europe. Resuscitation; 67:75-80 R. Busto, W.D. Dietrich, M.Y. Globus, et al. (1987). Small differences in intraischemic brain temperature critically determine the extent of ischemic neuronal injury J Cereb Blood Flow Metab, 7 pp. 729–738 F. Sterz, P. Safar, S. Tisherman, et al. (1991). Mild hypothermic cardiopulmonary resuscitation improves outcome after prolonged cardiac arrest in dogs Crit Care Med, 19 pp. 379–389 Zeiner A, Holzer M, Sterz F, et al. (2001). Hyperthermia after cardiac arrest is associated with an unfavourable

neurologic outcome. American Medical Association; 161:2007-12 6. Vargas M, Sutherasan Y, Servillo G, et al. (2015). What is the proper targeted temperature for out-of-hospital cardiac arrest? Elsevier; 29:425-434 7. Preberdy MA, Callaway CW, Neumar RW, et al. (2010). Post-cardiac arrest care: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation: 122:Suppl 3:S768-S786 8. Jerry P. Nolan, Jasmeet Soar, Alain Cariou, et al. (2015). European Resuscitation Council and European Society of Intensive care Medicine Guidelines for Post-resuscitation Care 2015. Section 5 of the European Resuscitation Council Guidelines for Resuscitation 2015. Elsevier Resuscitation: 202-222 9. Jennet B, Bond M, (1975). Assessment of outcome after severe brain damage. Lancet; 1:480-4 10. Nielsen N, Wetterslev J, Cronberg T, et al. (2013). Targeted temperature management at 33˚C versus 36˚C after cardiac arrest. N Engl J Med; 369:2197-206 11. Lopez-de-Sa E, Rey J.R, Armada E, et al. (2012). Hypothermia in comatose survivors from out-of-hospital cardiac arrest. Resuscitation science. Circulation Dec 11;

February 2018 | AMSj Vol. 10


17 SOLVING STATISTICS 126(24): 2826-33 12. Joo Kim J, Jun Yang H, Su Lim Y, et al. (2011). Effectiveness of each target body temperature during therapeutic hypothermia after cardiac arrest. Ajem: (29), 148-154 13. Holzer M, Cerchiari E, Martens P, et al. (2002). Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med (8); 346 14. Bernard SA, Gray TW, Buist MD, et al. (2002). Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med 346: 557-63 to Resuscitation Medicine, 3. London/ Philadelphia, WB SaundersCo: 261-268 15. Ajam K, S Gold L, S Beck S, et al. (2011). Reliability of the Cerebral Performance Category to classify neurological status among survivors of ventricular fibrillation arrest: a cohort study. Scand J Trauma Resusc Emerg Med.; 19: p38 16. Nielsen N, Wetterslev J, Cronberg T, et al. (2013). Targeted temperature management at 33°C versus 36°C after cardiac arrest. Supplementary Appendix. N Engl J Med; 369:2197-206. 17. Cummins O. R., Eisenberg M. S., Hallstrom A. P, et al. (1985). Survival of out-of-hospital cardiac arrest with early initiation of cardiopulmonary resuscitation. The American Journal of Emergency Medicine, 3, 114-119 18. Safar P, Bircher NG. (1988). Cardiopulmonary Cerebral Resuscitation: Basic and Advanced Cardiac and Trauma Life Support: An Introduction to resuscitation medicine, 3rd edn. Saunders, London, p 267 19. Cronberg T, Lilja G, Horn J, et al. (2015). Neurologic function and health-related quality of life in patients following targeted temperature management at 33˚C vs. 36˚C after out-of-hospital cardiac arrest: a randomized clinical trial. Jama Neurol; 72(6): 634-641 20. [20] Frydland M, Kjaergaard J, Erlinge D, et al. (2015). Targeted temperature management of 33˚C and 36˚C in patients with out-of-hospital cardiac arrest with initial non-shockable rhythm. Resuscitation 89: 142-148 21. Kjaergaard J, Nielsen N, Winther-Jensen M, et al. (2015). Impact of time to return of spontaneous circulation on neuroprotective effect of targeted temperature management at 33 or 36 degrees in comatose survivors of out-of-hospital cardiac arrest. Resuscitation 96: 310316

Help! How do I get a normal distribution? N. de Klundert, Master student, Academic Medical Center, Amsterdam R. Holman, Clinical Research Unit, Academic Medical Center, Amsterdam

Background

The distribution of a continuous numerical variable tells you something about how likely each possible outcome or groups of outcomes will occur. The distribution of a continuous numerical variable can be described as normal or non-normal. A normal distribution is the name for a specific mathematical concept that follows a symmetric bell shaped curve and is completely defined by its mean and standard deviation. Approximately 68% of the observations fall between one standard deviation below and one standard deviation above the mean. And approximately 95% of the observations fall between two standard deviations below and two standard deviations above the mean. Normal distribution is an important term within the field of statistics. If you have a normal distribution you can perform a certain number of statistical tests that you cannot perform if your distribution is non-normal.1

Question

I am currently conducting a study on the effects of medication on people with Alzheimer’s disease. Part of this study is the collection of a number of numerical values. I wanted to perform a multiple linear regression with this data but the problem is that one variable is not normal distributed. How do I solve this problem? Is it possible to still preform the statistical test that I want?

Answer

Normally the advice would be to just use statistical tests that were designed for data with a non-normal distribution, but since you want to perform a multiple linear regression, a non-normal distribution makes this more difficult. However, when performing linear regression, the

AMSj Vol. 10 | February 2018


18 SOLVING STATISTICS

the natural logarithm and different outcomes. Notice how the larger an outcome gets the lesser impact it has on the natural logarithm and thus creating a workable data set. The observed outcome is on the x (horizontal) axis and the natural logarithm transformed outcome on the y (vertical) axis. An original value of 2 is transformed to a value of 0.69 and an Figure 1 Data before and after a natural logarithmic transformation

actual assumption is that the residuals will follow a normal distribution. Even if your variables do not follow a normal distribution, the residuals may do so. Hence, you can perform the linear regression analysis and then examine whether they follow a normal distribution. If there are extreme values, known as outliers, in your data, they can skew the distribution to the left or right. For example, incorrect or extreme values may have a great effect on the distribution. You can try and see what the distribution will be if these values are eliminated. However, remember that you have to explain why it is acceptable to delete a value from your data. You should not just delete them because it makes your analysis easier! A second way to solve your problem is to transform your data. A non-normal distribution may become closer to a normal distribution if the values are transformed using a mathematical formula. The logarithmic transformation is often used. For example, if your values are 12, 18, 27, 35 and 85 their natural logarithm values would be 2.48, 2.89, 3.30, 3.56 and 4.44. As you can see, these numbers are much closer together. For example, if you take a look at FIGURE 1, you can see that there is a non-normal distribution when we look at the data. However, when we apply a natural logarithm it becomes a normal distribution that follows the symmetric bell curve. In FIGURE 2 you can see the association between

original value of 8 is transformed to a value of 2.08. You can also use the square root (√x) transformation. If you use a transformation, you may have to perform a back transformation to enable you to interpret the results of your statistical analysis.

Figure 2 The natural logarithmic transformation of the outcome

Reference

1.

Petrie A, Sabin C. Medical Statistics at a glance. West sussex, UK: Wiley Blackwell; 2009.

February 2018 | AMSj Vol. 10

Read a paper, but question the statistical analyses? Let us know via AMSj.nl or our Facebook page!


19 INTERVIEW

Interview prof. dr. Ulrich Beuers Interviewed by J. Saris

Area of expertise: Cholestatic and autoimmune liver and bile duct diseases. What are your research interests? The role of a ‘biliary bicarbonate umbrella’ for the pathogenesis and effective treatment of various chronic progressive hepatobiliary diseases. The pathophysiology and treatment of itch as a major debilitating symptom in cholestasis. The pathogenesis and diagnosis of an only recently defined systemic autoimmune disorder, IgG4-related disease, which closely mimics other benign and malignant hepatobiliary and pancreatic diseases. Publications: Numerous. Awards: Numerous national and international awards/acknowledgements amongst others three German scientific awards, two member of honour awards (Czechia and Slovakia), PSC award and EASL mentor. When did you decide to become a physician? As a child, I had grown up on a ‘Zauberberg’ (Thomas Mann) next to a hospital for pulmonary diseases - my father was internist-pulmonologist and thoracic surgeon - in the forests of the Sauerland not far from a farm, a kind of a paradise for my brothers and me. Before the age of 18, I had excluded medicine and had looked for different options. But at 18, I underwent a mind-switch, when I recognized that economy, law, engineering or professional music were not my real passions. I turned 100% convinced that I had to become a caring physician. Can you tell us more about your studies? Since we did not reach the scores on our oldfashioned Gymnasium which were given in the neighbouring schools, I knew that I had to go abroad if I wanted to study medicine. I had an opportunity to start in Gent/Belgium, where I

Curriculum vitae 1956 Born 1983 Medicine: University of Freiburg, Germany 1983 Dissertation: University of Freiburg: “Die Wirkung des Renin-Angiotensin Aldosteron-Systems auf die Freisetzung von ß-Endorphin- und alfa-Lipoprotein Immunreaktivität in der wachen Ratte” 1984-86 Post-doc Biochemistry in Göttingen, Germany 1986-91 And 1993-95 Specialization Internal Medicine, Gastroenterology & Hepatology in Munich, Germany 1991-93 Research at Yale, New Haven, United States of America 1994 Habilitation: “Zu klinischer Wirksamkeit, Metabolismus und Wirkmechanismus von Ursodeoxycholsäure bei cholestatischen Leberkrankheiten” 1995-02 Oberarzt, Internal Medicine, Gastro enterology & Hepatology, Munich 2001 Professor 2003-06 Leitender Oberarzt, Internal Medicine, Gastroenterology & Hepatology, Munich Current positions: Core Professor of Gastroenterology and Hepatology at the AMC in Amsterdam, Head of Hepatology, Program director of Gastroenterology and Hepatology, Chairman of the national education committee for Gastroenterology and Hepatology, Chairman of the Netherlands Association for the Study of the Liver, Associate Editor, Journal of Hepatology.

AMSj Vol. 10 | February 2018


20 INTERVIEW

successfully passed the 3 years of preclinical studies, an enormous challenge for a foreigner! Thereafter, I continued my clinical studies in West-Berlin, Germany (monthly highlight: Berlin Philharmonic Orchestra) and after one year I moved to Freiburg where I received my ‘Approbation’ (bul) as medical doctor. Do you notice any differences between the German, Belgian and Dutch medical education? I think that German and Dutch medical schools are well comparable, and that the average medical knowledge of Dutch and German students at the end of the studies is also comparable, but that Dutch and German students may be behind at the average Belgian student when considering basic knowledge. In my view, the Dutch system is particularly strong in the well-organized specialist training, at least for my discipline, Gastroenterology & Hepatology. Did you perform any research related activities while studying? As a student, I had for a while the plan to become a ‘third world doctor’. I very much enjoyed 3- and 4-month medical clerkships on a faraway Indonesian island and in Northeastern Brasil. During the medical studies in Freiburg I started my dissertation on the field of neuropharmacology working for about 18 months on ‘The effect of the Renin-Angiotensin-Aldosterone system on β-endorphin and α-lipoprotein immunoreactivity in the rat’. I had chosen for experimental work in order to make this experience at least once in my lifetime. I was lucky to find an enthusiastic mentor (most important!) and was later unexpectedly awarded for this thesis.

‘‘I am seriously ‘infected’ by the challenge to do clinical work in combination with research.’’

Professors in Internal Medicine was my role model. He was a most respected chairman and highly gifted clinician, teacher and researcher on the field of hepatology. He convinced me to do a twoyear post-doc period in Biochemistry in Göttingen to study liver metabolism before entering the clinic. Thereafter I joined the Department of Medicine, Gastroenterology and Hepatology in Munich and was mentored by Professor Paumgartner. I had been seriously ‘infected’ by the challenge to do clinical work in combination with research. In Munich, I combined my first clinical trials in cholestatic liver diseases with experimental work. After almost 6 years, I interrupted my clinical training and went to Yale University School of Medicine for experimental studies to further substantiate a hypothesis on which I worked.

‘‘I can highly recommend students and fellows to study and work abroad in order to gain experience.’’ I can highly recommend students and fellows to study and work abroad in order to gain experience. Back in Munich, I finished my training in Internal Medicine, Gastroenterology and Hepatology, became an associate professor and finally professor in Internal Medicine. After 20 years, I felt that I need a new challenge. The AMC in Amsterdam appeared to me the most attractive and challenging among the offers I received. I came to Amsterdam in 2007. What are your hobbies? I very much enjoy sports (ski, football, tennis, squash, sailing, biking), classical music, reading and spending time on our island on the Vinkeveense plassen.

When did you realize you wanted to specialize in Gastroenterology and Hepatology? Professor Gerok, one of Feiburgs core February 2018 | AMSj Vol. 10


21 subject 101

Glucosuria – trick or treatment? M.J.B. van Baar

The color and odor, quantity, clarity & viscosity, foaminess, saltiness & sweetness of one’s urine was once ample information to determine the flaws in our personal well-being, summarized in the acronym of a modern shoe; ASICS. Anima sana in corpore sano. Both soul and body were represented in our clear-golden liquid.1 A particular disease, diagnosed spot-on through this method, is one of the oldest medical entities ever described.2 Already in 1500 AD, an Egyptian manuscript mentions an epithetical name; too great emptying of the urine. This emptying, defined in 230 AD by the Greek Apollonius of Memphis as διαβήτης (diabetes), “to pass through” (dia – through, betes – to go), was sufficient to characterize the condition for centuries.3 Only in 1675, when Thomas Willis, known for his discovery of the arterial circle in our brain, coined in his Pharmaceutice rationalis the term mellitus, “honey-like”, to differentiate between mellitus and insipidus, “tasteless”, the name diabetes mellitus emerged as a clinical condition.4 The doctor’s trick for diagnosis was simply to taste the urine. The question is, where does this sweetness of one’s urine come from? Although small enough to pass freely through the glomerular barrier, glucose is under normal conditions almost absent in urine. Given the fact that approximately 180 L of plasma is filtered every 24 hours, and given the fact that a mean plasma glucose concentration is about 100 mg/dL (5.6 mmol/L), the amount of glucose filtered every day is more or less 180 g, though subsequently it is entirely reabsorbed and returned into the blood plasma.5 An increase in tubular glucose concentration is paralleled by an increase in glucose reabsorption up to approximately 11 mmol/L, and as such, no glucose is present in urine.6 The symporters responsible for this reabsorption are two sodium-glucose cotransporters (SGLTs), of which both are osmotic gradient dependent symporters that couple sodium to glucose reabsorption in a 1:1 (SGLT-2) or 2:1 (SGLT-1) ratio. The two are located sequential-

ly in segments the proximal tubule; SGLT-2 is situated in the first segment (S1) and SGLT-1 in the adjacent two segments (S2/S3) (see FIGURE 1).7 Roughly 90% of the glucose in the ultrafiltrate is reabsorbed through the high-affinity low-capacity SGLT-2, while the remaining 10% is reabsorbed through the low-affinity high-capacity SGLT-1.7 When the amount of glucose filtered exceeds the maximal reabsorption capacity, also called the renal threshold, glucosuria occurs. In diabetes mellitus, paradoxically this renal threshold is raised as a result of circa 30% increase of glucose reabsorption, probably due to a corresponding upregulation of SGLT-2, which adds to the persistence of a hyperglycemic state.8,9 Although glucosuria has become obsolete as diagnostic test due to modern day laboratory tools, it can still be used as an indication of early or inadequately controlled diabetes mellitus. For both hygienic and personal reasons, doctors today are more eye-orientated and will avoid this tasty task for science and prefer to use colorimetric alternatives such as the urine strip to measure the amount of glucose in urine. Nonetheless, through the development and implementation of a new class of antihyperglycemic agents for type 2 diabetes patients, the sweetness of urine has regained clinical attention. The upregulation of SGLTs in the proximal tubule in humans with impaired glucose metabolism have led to the idea that these transporters could be a therapeutic target for glycemic control. Observations in the early 1980s indicated that phlorizin (which occurs naturally in certain parts of plants such as the root bark of an apple tree) was a potent inhibitor of both SGLT-1 and SGLT-2. Through its

AMSj Vol. 10 | February 2018


22 subject 101

FIGURE 1 Sodium-glucose cotransporters (SGLTs) responsible for reabsorption of glucose

major affinity with SGLT-2, the substance proved to be a useful insulin-independent antihyperglycemic drug as a result of the inhibition of renal glucose reabsorption, without causing hypoglycemia. However, phlorizin turned out to be unsuitable for clinical use, because of its minor affinity with SGLT-1, present in the small intestine, where it enables absorption of glucose and galactose from food.10 Potent inhibition of SGLT-1 in the gut leads to intolerable gastrointestinal adverse effects due to reduced intestinal glucose absorption and subsequent osmotic diarrhea. For this reason, less potent SGLT-1 inhibitors have recently been developed, to exploit the antihyperglycemic benefits of inhibition of gut glucose absorption without the adverse effects.11 Likewise, specific SGLT-2 inhibitors have been developed, of which three are currently approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for patients with type 2 diabetes; canagliflozin, dapagliflozin and empagliflozin, which are all acceptable second- or third-line options in antihyperglycemic treatment.12 Through their practical and also promising properties, indicated by the EMPA-REG Outcome Trial, where empagliflozin showed long-term cardiovascular and renal benefits beyond glucose control, SGLT inhibition might become one of our future doctor’s top-notch tricks in treatment of diabetes.5,13

Acknowledgements

Special thanks to E. van Bommel and M.H.A. Muskiet, research physicians at Diabetes Centre of the VUmc for their knowledge on the subject and the use of figures.

References

1. 2. 3. 4.

5.

6. 7.

8.

9.

10. 11.

12. 13.

Nicholson JK, Lindon JC. Systems biology: Metabonomics. Nature. 2008;455(7216):1054-6. Leutholtz BC, Ripoll I. Exercise and Disease Management. CRC Press; 2011. p. 25. Zajac J, Shrestha A, et al. Principles of Diabetes Mellitus. Springer Science & Business Media; 2010. p. 3-16. Willis T. Pharmaceutice Rationalis, sive Diatriba de Medicamentorum Operationibus in humano Corpore 1675. Available from: https://books.google.nl/books?id=Kg49AAAAcAAJ&redir_esc=y. van Bommel EJ, Muskiet MH, Tonneijck L, et al. SGLT2 Inhibition in the Diabetic Kidney-From Mechanisms to Clinical Outcome. Clin J Am Soc Nephrol. 2017;12(4):700-10. Barrett KE, Barman SM, Boitano S, et al. Ganong’s Review of Medical Physiology. 21st ed: McGraw-Hill Companies; 2003. p. 702–30. Rieg T, Masuda T, Gerasimova M, et al. Increase in SGLT1-mediated transport explains renal glucose reabsorption during genetic and pharmacological SGLT2 inhibition in euglycemia. Am J Physiol Renal Physiol. 2014;306(2):F188-93. DeFronzo RA, Hompesch M, et al. Characterization of renal glucose reabsorption in response to dapagliflozin in healthy subjects and subjects with type 2 diabetes. Diabetes Care. 2013;36(10):3169-76. Rahmoune H, Thompson PW, Ward JM, et al. Glucose transporters in human renal proximal tubular cells isolated from the urine of patients with non-insulin-dependent diabetes. Diabetes. 2005;54(12):3427-34. Ehrenkranz JR, Lewis NG, Kahn CR, et al: a review. Diabetes Metab Res Rev. 2005;21(1):31-8. Sands AT, Zambrowicz BP, Rosenstock J, et al. Sotagliflozin, a Dual SGLT1 and SGLT2 Inhibitor, as Adjunct Therapy to Insulin in Type 1 Diabetes. Diabetes Care. 2015;38(7):1181-8. ADA. Standards of medical care in diabetes--2016. Diabetes Care 2016;39 Suppl 1 (Jan 2016):S72–74. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373(22):2117-28.

February 2018 | AMSj Vol. 10


23 ARTICLE

Adoptive cell therapy using tumor infiltrating lymphocytes versus marrow infiltrating lymphocytes in the treatment of cancer: a systematic-review M. de Jong, VU University Medical Center, Amsterdam

Abstract

B a c kg r o u n d Adoptive cell therapy (ACT) of tumor-infiltrating lymphocytes (TILs) is a powerful method of immunotherapy that has great potential to treat patients with metastatic melanoma and patients with other solid tumors. ACT using TILs is now in various early- and late-stage clinical promising trials. Recently, research has shown that the use of ACT using marrow-infiltrating lymphocytes (MILs) could lead to regression of cancer in multiple myeloma (MM) patients. This systematic review outlines the differences between these two ACTs with the aim to pinpoint the areas of research for the therapeutic use of MILs in the future. The main intent of this review is to investigate the future outlook of MILs and to investigate whether a future transition from ACT using TILs to ACT using MILs in the treatment of cancer could emerge. M e t h o d s Reporting of this review was done systematically. A systematic search was conducted in the database of PubMed. Literature on ACT using TILs and MILs was examined. A total of 14 articles were included in this review. One article compared the neurologic function and mortality of targeted temperature management (TTM) at 33˚C and 36˚C, but concluded no difference in outcome between 33 and 36C. The other four articles examined a targeted temperature between 32˚C and 37˚C. All four articles supported therapeutic hypothermia at 33/34˚C. The CPC 1-2 and mortality of 33˚C did not show high discrepancy compared to 36˚C. RESULTS Clinical trials until now have demonstrated that the use of MILs leads to cancer regression. The easy accessibility of the bone marrow, the absence of the need for surgical removal, the short extension time and the 100% successful collect and expand rates are main advantages of ACT using MILs compare to ACT using TILs. c o n c l u s i o n Whether a transition from ACT using TILs to ACT using MILs will emerge in the future depends on the outcome of future trials using MILs in the treatment of various solid tumors.

INTRODUCTION

Adoptive T cell therapy

Over the past years, immunotherapy has made a significant contribution in the treatment of many malignancies. The primary goal of anti-cancer immunotherapy is to use the immune system for recognizing and destroying tumor cells. One of the main treatment applications within cancer immunotherapy has been adoptive T-cell therapy. TILs

Adoptive cell therapy (ACT) using tumor infiltrating lymphocytes (TILs) uses tumor-reactive T cells that are already present within the tumor. These TILs are obtained from the patient’s own tumor tissue and after ex-vivo activation and expansion re-infused.1 ACT using TILs has been used in promising clinical trials to treat metastat-

ic melanoma and some other solid tumors, like breast-, ovarian- and renal cell cancer.1-4 MILs

In multiple myeloma, a hematopoietic malignancy of the antibody-producing plasma cells, the bone marrow is the tumor microenvironment. Noonan et al. 2005 showed that the tumor-specific T-cells obtained from the tumor microenvironment in multiple myeloma demonstrate high tumor specificity after activation and expansion.5 Recently in 2015, Noonan et al. have shown that the use of these marrow-infiltrating lymphocytes (MILs) for ACT could lead to tumor regression in multiple myeloma.6 MILs appear to have a lot of unique features that make them an ideal source of T cells for ACT. They are currently being used in several different clinical trials.7

AMSj Vol. 10 | February 2018


24 ARTICLE

FIGURE 1 Flowchart of the search strategy

The intent of this systematic review is to investigate whether a possible future transition from ACT using TILs to the new approach of ACT using MILs in the treatment of cancer could occur. Despite existing research, a systematic review concerning ACT using TILs and MILs is missing. Therefore, the primary aim of this review is to compare and contrast various aspects of ACT using MILs and TILs. In addition, thereby identifying the areas of research for the therapeutic use of MILs in the future. To this end, I performed a comprehensive research of existing literature on immunotherapy using TILs and MILs.

METHODS

On the 13th of January 2017 an English literature research was conducted in PubMed to identify relevant articles addressing ACT using TILs and MILs. The database search began with the search for articles addressing ACT using MILs. This search was conducted in PubMed using the following search term: Marrow infiltrating lymphocytes and adoptive cell therapy. The following Mesh terms were used: “Lymphocytes, Tumor-Infiltrating” and “Bone Marrow”. Studies addressing ACT using MILs in the abstract or title were selected for inclusion. Exclusion criteria were articles not published in English and reviews.

The reference lists of the retrieved studies were reviewed to identify articles that could also be selected for inclusion, but that did not show in the database search. The search for articles addressing ACT using TILs was conducted in PubMed using the search terms: tumor infiltrating lymphocytes, adoptive cell therapy and clinical trial. The following Mesh terms were used: “Lymphocytes, Tumor-Infiltrating” and “Immunotherapy, Adoptive”. Studies addressing ACT using TILs in the abstract or title were selected for inclusion. Exclusion criteria were: reviews and articles not published in English. After checking reference lists, two additional articles were included. The first and second search together yielded a total of 14 articles. These search strategies are shown in FIGURE 1.

RESULTS

ACT using TILs has a long clinical history with multiple clinical trials.1-3,8 MILs offer a new approach to ACT and have some benefits with respect to TILs. An historical overview of the discovery and development of ACT using TILs and MILs is summarized in FIGURE 2. An overview of the differences between ACT using TILs and using MILs will be presented in this section.

February 2018 | AMSj Vol. 10


25 ARTICLE Culture and expansion: TILs vs. MILs First of all, not all patients with solid tumors have TILs that can be expanded. In the trial of Rosenberg et al. 2011 only 67% of all patients had reactive specific TILs that could be identified.4 In Tran et al. 2008 TIL cultures could be prepared in only 80% of tumor specimens.9 From the 19 patients that were enrolled in the study in of Pilon-Thomas et al. 2015, TILs were successfully grown from the tumors of only 17 patients (89%).3 In contrast, Noonan et al. 2015 showed that the MILs attempts to collect and expand have been successful in 100% of patients.6 All the 21 patients from this first clinical phase I trial had MILs that were obtainable. In Domschke et al. 2013 10 all 16 included metastatic breast cancer patients showed tumor-reactive bone marrow T cells at the beginning of the study. Up to now, MILs have been successfully obtained and expanded in all patients enrolled thus far.4,5 Secondly, the standard method for producing TIL cultures for re-infusion requires ex vivo expansion in IL-2 and identification of tumor reactive cells by immunological assays. This TILs expansions protocol is lengthy. The time and expertise involved make large-scale clinical translation of this approach challenging. MILs, on the other hand, require a much shorter duration of culture time. MILs expansions take seven days in total whereas TILs expansions have a mean age of 25 days.4,6,9 This shortens preparation time leading to a reduced dropout rate. The long expansion time of the TILs cultures have led to the use of Young-TILs in 2008 by Tran et al.9 The YoungTILs expansion method has a mean age of 12 days, still longer than the MILs expansion time of seven days. Accessibility: TILs vs. MILs

Another major difference between TILs and MILs is the way in which they are obtained. TILs are harvest by a surgical procedure, which limits the general applicability. In the clinical trials using TILs only patients were included with a metastatic nodule of at least 2 cm diameter that was suitable for resection.2-4,9 This means that not all patients with metastatic melanoma can receive the TILs treatment. In contrast, MILs are easily accessible from the bone marrow with a simple bedside procedure.6 The ease of obtaining MILs

and the absence of a surgery provide major advantages over the use of TILs. Antigen specificity: TILs vs. MILs A further difference between TILs and MILs is the antigen specificity. MILs that induce anti-tumor activity show to recognize proteins on the surface of tumor cells, which we called tumor associated antigens (TAAs).11,12 Trials showed that MILs target a broad range of these TAAs present on the surface of both mature terminally differentiated plasma cells as well as their precursors.5,6 In contrast to the surface TAAs recognition of the MILs therapy, TILs therapy targets neo-antigens.2,10,13,14 The treatment response of TILs therapy in melanoma patients is high because melanoma is a highly mutated form of cancer, which means the presence of a lot of neo-antigens that can be frequently recognized by TILs.1 A disadvantage of the neo-antigens recognition of the TILs is that tumors with low mutational burden are not suitable for TILs therapy. The reason for this is that tumors with a low mutational burden may express less neo-antigens.

DISCUSSION

MILs are cells that have been shaped by the bone marrow, which have essential properties that make them an ideal candidate for ACT. However, it is not yet possible to say if a transition from ACT using TILs to ACT using MILs will emerge in the future as a result of several limitations. First of all, it is difficult to compare the results of individual studies of ACT using TILs and ACT using MILs directly. There are no clinical trials in which the effect of ACT using TILs and ACT using MILs are directly compared with each other. The MILs are only compared with ACT using PBLs. Therefore, it is impossible to conclude whether MILs transfer will have a better or equal anti-cancer effect compared to TILs transfer. Besides this, in contrast to TILs therapy, little is known about the long-term effects, the optimal dose and the optimal time to start the MILs therapy. Further research is needed to address these points. In addition, the use of MILs in clinical trials focuses on patients with MM and on patients with

AMSj Vol. 10 | February 2018


26 ARTICLE

FIGURE 2 Timeline | A brief history of the development of ACT using TILs and MILs

metastatic breast cancer.10 The transfer of TILs is mainly tested on patients with metastatic melanoma but trials on patients with other solid tumors, like ovarian-, renal cell- and breast cancer, are ongoing.1-4,6 Further studies should be done to investigate the ability to utilize MILs in ACT in various other solid tumors. Next to these limitations, there are some missing variables in the research of MILs transfer. First of all, trials including MILs therapy in combination with other therapies are missing. Checkpoint antibodies blocking PD-1 or its ligand PD-L1 have been researched in combination with ACT using TILs and have shown promising results in renal cell carcinoma and ovarian carcinoma.1 The combination of TILs therapy and these checkpoint inhibitors result in a higher anti-tumor effect. Anti-PD-L1 antibody after ACT can also enhance the persistence of TILs in melanoma murine model. However, more research with this combination therapy is required.3 In contrast, no research has been done concerning MILs therapy in combination with checkpoint antibodies. Further research is necessary to see whether checkpoint inhibition can also have a positive effect on the MILs transfer.

A second missing variable in the research of MILs transfer is the correlation with the telomere length of the infused cells. A strong correlation between the probability of a response and the telomere length of the re-infused TILs has been described.1,4,9 This suggests that the telomere length of the TILs is an important aspect of their capability to cause tumor regression in patients. Whether the telomere length also has a correlation with the efficacy of the MILs therapy has yet to be investigated. This meta-analysis is also subjected to some limitations. The numbers of included studies is small, and only the database from PubMed is used. This meta-analysis contains a degree of publication bias since only trials published in English were included.

CONCLUSION

In conclusion, MILs therapy show promising effects in the treatment of MM and future research has to show whether the transfer of MILs has the same effect on solid malignancies as the transfer of TILs. MILs differ from TILs in that they come from different compartments and recognize different tumor antigens. The easy

February 2018 | AMSj Vol. 10


27 ARTICLE

accessibility of marrow, the absence of the need for surgical removal, the short extension time and the 100% successful collect and expand rates are major advantages of ACT using MILs as compared to TILs. Accordingly, no conclusions can be drawn on the overall future outlook of ACT using MILs until more evidence about the effectiveness is available.

Acknowledgments

I would like to acknowledge Dr Tuna Mutis (Associate professor VU University Medical Center) for his supervision and feedback.

REFERENCES

11. Feuerer, M., Beckhove, P., Bai, L., et al. (2001). Therapy of human tumors in NOD/ SCID mice with patient-derived reactivated memory T cells from bone marrow. Nature Medicine, 7(4), pp. 452–458. 12. Di Rosa, F. & Santoni, A. (2002). Bone marrow CD8 T cells are in a different activation state than those in lymphoid periphery. European Journal of Immunology, 32(7), pp. 1873–1880. 13. Muul, L. M., Spiess, P. J., Director, E. P., et al. (1987). Identification of specific cytolytic immune responses against autologous tumor in humans bearing malignant melanoma. The Journal of Immunology, 138(3), pp. 989-995. 14. Rosenberg, S. A., Spiess, A.,& Lafreniere, R. (1986). A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science, New Series, 233(4770), pp. 1318-1321.

1.

Andersen, R., Donia, M., Westergaard, M. C. et al. (2015). Tumor infiltrating lymphocyte therapy for ovarian cancer and renal cell carcinoma. Human Vaccines & Immunotherapeutics, 11(12), pp. 2790–2795. 2. Dudley, M. E., Wunderlich, J. R.,1 Robbins, P, et al. (2002). Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science, 298(5594), pp. 850-854. 3. Pilon-Thomas, S., ,Kuhn, L., Ellwanger, S., et al. (2012). Efficacy of adoptive cell transfer of tumor infiltrating lymphocytes after lymphopenia induction for metastatic melanoma. Journal of Immunotherapy, 35(8), pp. 615–620. 4. Rosenberg, S. A., Yang, Y. C., Sherry, R. M., et al. (2011). Durable complete responses in heavily pretreated patients with metastatic melanoma using T cell transfer immunotherapy. Clinical Cancer Research, 17(13), pp. 4550–4557. 5. Noonan, K. A., Matsui, W., Serafini, P., et al. (2005). Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors. Cancer research, 65(5), pp. 2026–2034. 6. Noonan, K. A., Huff, C. A., Davis, J., et al. (2015). Adoptive transfer of activated marrow-infiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma. Science Translational Medicine, 7(288), pp. 278-288. 7. Noonan, K. A. & Borello, I. (2015). Marrow Infiltrating Lymphocytes: Their Role in Adoptive Immunotherapy. The Cancer Journal, 21(6), pp. 501-505. 8. Rosenberg, S. A., Packard, B. S., Aebersold, P. M., et al. (1988). Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. The New England Journal of Medicine, 319, pp. 1676-1680. 9. Tran, K. O., Zhou, J., Durflinger, K. H., et al. (2008). Minimally cultured tumor-infiltrating lymphocytes display optimal characteristics for adoptive cell therapy. Journal of Immunotherapy, 31(8), pp. 754-751. 10. Domschke, C., Ge, Y., Bernhardt, I., Set al.(2013). Longterm survival after adoptive bone marrow T cell therapy of advanced metastasized breast cancer: follow-up analysis of a clinical pilot trial. Cancer Immunology Immunotherapy, 62(6), pp. 1053–1060.

AMSj Vol. 10 | February 2018


28 ARTICLE

Hyperthermic intraperitoneal chemotherapy for patients with peritoneal metastases of colorectal cancer treated with adjuvant chemotherapy: A systematic review Y. Salhi, medical student Department of Surgery, VU University Medical Centre, Amsterdam Supervised by: dr. J.B. Tuynman, colorectal surgeon, N.R. Sluiter, PhD candidate Department of Surgery, VU University Medical Centre, Amsterdam, The Netherlands

Abstract

B a c kg r o u n d Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) is nowadays the preferred treatment for patients suffering from isolated peritoneal carcinomatosis (PC) of colorectal cancer (CRC). CRS is resection of all visible macroscopic tumor deposits in the abdomen and HIPEC is flushing of the abdomen with heated chemotherapy, usually Mitomycin. Despite its potential efficacy and effectiveness, this treatment is still associated with relatively high mortality and morbidity rates. Therefore, accurate selection of patients who will benefit from this treatment is warranted. We hypothesized that development of PC despite treatment with adjuvant chemotherapy for the primary tumour might have a negative influence on outcome after CRS and HIPEC. This review was performed to compare the prognosis of HIPEC patients treated with adjuvant chemotherapy for their primary colon carcinoma to the prognosis of patients that not receive adjuvant chemotherapy. M e t h o d s A systematic literature search was conducted according to the PRISMA guidelines to identify studies reporting HIPEC patients that received adjuvant chemotherapy for their primary disease. From the 259 identified articles, eight were considered suitable for this review. Survival data of patients who received adjuvant chemotherapy for their primary carcinoma prior to CRS and HIPEC were compared to survival data of patients who did not receive adjuvant therapy. Furthermore, morbidity rates were compared between these two groups of patients. R e s u lt s From the eight included studies, six studies did not report an effect of adjuvant chemotherapy on survival after CRS and HIPEC. Two studies reported that worse median survival was associated with treatment with adjuvant therapy (19.2 vs 20.4 months, P=0.01 compared to 31 vs 45 months, P=0.008). One study reported that a better median survival was associated with treatment with adjuvant therapy (40% vs 15%, P=0.040). C o n c l u s i o n Currently, there is no evidence for excluding patients that developed PC regardless of adjuvant chemotherapy from HIPEC treatment. However, large prospective studies are required to investigate whether adjuvant chemotherapy is associated with survival of CRC patients after CRS and HIPEC. a bb r e v i at i o n s CRC = Colorectal cancer PC Peritoneal carcinomatosis, CRS = Cytoreductive surgery, HIPEC = Hyperthermic intraperitoneal chemotherapy, ACT = Adjuvant chemotherapy, PRISMA = Preferred Reporting Items for Systematic Reviews and Meta-Analyses, ASCO = American Society of Clinical Oncology, OS = Overall survival, NR = Not Reported in the article, USA = Univariate Survival Analysis, MSA = Multivariate Survival Analysis, CI = Confidence Interval

INTRODUCTION Rationale

Colorectal cancer (CRC) is the second most common cancer in the Netherlands and the third most common cancer worldwide.1,2 According to the American Society of Clinical Oncology (ASCO) guideline, patients with high risk stage II

and stage III colon cancer should receive adjuvant chemotherapy, (most commonly Capecitabine/ Oxaliplatin (CAPOX) or a combination of Leucovorin, 5-fluorouracil and Oxaliplatin (FOLFOX)) to minimise the risk on residual disease, local recurrence or distant metastases.3

February 2018 | AMSj Vol. 10


29 ARTICLE

Following the liver, the peritoneum is the second most common site of metastases originating from CRC. Approximately 15% of all patients develop synchronous peritoneal metastases and 25% of the patients develop metachronous peritoneal carcinomatosis (PC).4,5,6 A selected group of patients with isolated PC is treated locally with cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC), resulting in a 5-year survival rate equal to that of patients undergoing resection for colorectal liver metastases (35-45%) and a median survival of 33 months.7-11 Despite the improved survival rates compared to survival rates after treatment with systemic chemotherapy, treatment with CRS and HIPEC has mortality rates of 5% and morbidity rates of 15-18%, regardless the extent of the disease, which makes an accurate and careful selection of patients who will benefit from this treatment of great importance.7,9,12,13 Criteria for selecting patients with PC are isolated and limited PC (a maximum of 5 out of 7 regions affected in the abdomen). Patients with distant metastases are excluded to undergo CRS and HIPEC.14 However, selection of patients who meet this criteria is not always adequate because currently used imaging techniques have low sensitivity.15 Therefore, a good understanding of other patient related factors influencing outcome, and thus possibly improving selection, of patients with peritoneal metastases (PM) from CRC is of great importance. Probably more than 50% of HIPEC patients are treated with adjuvant chemotherapy for their primary colon carcinoma. Despite this adjuvant therapy, more than 25% develop isolated metastases to the peritoneum. We hypothesize that patients who develop PC despite adjuvant therapy might have more aggressive tumours with a poorer response to systemic chemotherapy and the combined treatment of CRS and HIPEC. Knowledge on this group of patients might help in deciding whether these patients should be treated with CRS and HIPEC. Objectives

The primary aim of this review is to investigate the added value of CRS and HIPEC in CRC

patients that developed PC despite treatment with adjuvant chemotherapy for their primary tumor. For this purpose we searched for literature that compared survival rates of HIPEC patients that received adjuvant chemotherapy before the HIPEC procedure to HIPEC patients that did not receive adjuvant chemotherapy. The hypothesis is that patients who develop PC despite adjuvant therapy have a poorer prognosis compared to those who develop PC without treatment with adjuvant chemotherapy. This knowledge could be of great value in the patient selection for the HIPEC procedure.

METHODS

This systematic review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement using the checklist. This statement contains a minimum set of concepts for systematic reviews. The PRISMA statement is an evidence-based guideline that is used worldwide.16 Information sources

A systematic search using the National Library of Medicine (MEDLINE/PubMed) was performed to identify publications on the prognosis of patients that developed progressive metachronous PC regardless of treatment with adjuvant chemotherapy for their primary CRC compared to patients with progressive metachronous PC who did not receive adjuvant therapy. Search strategy

The National Library of Medicine was searched using the medical subject headings (MeSH): peritoneum, neoplasms and adjuvant chemotherapy. We combined these MeSH terms by identifying the specific entry terms for each MeSH term. An important entry term for the MeSH term peritoneum was peritoneal, for the MeSH term neoplasms we identified the terms tumor, cancer and malignancy. The entry term identified for the MeSH term adjuvant chemotherapy was adjuvant drug therapy. After this, we combined all terms with Boolean operators: ((“Neoplasms”) OR (neoplas* OR tumor OR tumors OR malignanc* OR cancer) AND ((peritoneal OR peritoneum) OR (‘“Peritoneum”)) AND ((“Chemotherapy, Adjuvant”) AND (chemothera-

AMSj Vol. 10 | February 2018


30 ARTICLE ((“Neoplasms”) OR (neoplas* OR tumor OR tumors OR malignanc* OR cancer) AND ((Peritoneal OR peritoneum) OR ((“peritoneum”)) AND ((“Chemotherap, Adjuvant*) AND (chemotherapy OR drug AND therapy) AND adjuvant) Identification 259 articles

Screening

Eligibility

Included

256 excluded after title and abstract screening 3 articles

5 included after cross referencing (snowball search)

8 articles

FIGURE 1 PRISMA flowchart for inclusion of the studies16

py OR drug AND therapy) AND adjuvant). Using the previously described search strategy, we identified a total of 259 articles. The first step in our selection process was title and abstract screening to exclude irrelevant articles. Using the predetermined inclusion and exclusion criteria three articles were selected. The second step in the selection process consisted of cross-referencing of the obtained articles, which resulted in five extra articles. FIGURE 1 describes the literature search and the selection process. Study selection

Studies were selected based on predetermined inclusion and exclusion criteria described below. Inclusion criteria

Full-text studies in English investigating the prognosis of patients with isolated PC who underwent systemic adjuvant chemotherapy before CRS and HIPEC for their primary colon carcinoma were included. The articles should contain information of patient characteristics, tumor characteristics and treatment type. Exclusion criteria

Studies investigating the presence of distant metastases other than PC without reporting data from PC patients separately and studies investigating PC from non-colorectal primary tumours were

excluded. We also excluded studies investigating synchronous PC originating from CRC and the use of biologicals as adjuvant therapy instead of adjuvant chemotherapy for the primary carcinoma. If patients received neoadjuvant treatment before the surgical resection of the primary carcinoma, they were excluded as well. In addition, we only selected studies reporting on patients older than 18 years of age. Finally, we did not include case reports, meta-analyses, systematic reviews and articles written in other languages than English or Dutch. Data collection process

From the selected studies, we extracted the publication year, first author of the article, study design, total number of patients, percentage males, percentage females and the median age of the participants included in the study, adjuvant chemotherapy received for the primary tumour and survival data. If reported in the article, we extracted morbidity data as well.

RESULTS

All outcomes extracted from the studies are shown in Table 1, 2, 3 and 4. Table 1 provides an overview of the studies and main baseline characteristics. Table 2 shows survival data extracted from the eight articles. Table 3 is an overview of the overall morbidity and mortality rates for all patients after CRS and HIPEC. Table 4 shows

February 2018 | AMSj Vol. 10


31 ARTICLE Article no.

Study design

n

Male (%)

Female (%)

Median age (years)

No. of patients treated with ACT (%)

No. of patienst not treated with ACT (%)

Median follow up (months)

1.Glehen Aet al. (2004)

Retrospective cohort study

506

46.1%

53.9%

51

275 (54.3%)

231 (45.7%)

53

2. Shen et al. (2008)

Prospective cohort study

121

51%

49%

54

40 (72.2%)

15 (27.3%)

86

3. Glehen et al. (2010)

Retrospective cohort study

1290

43.5%

56.5%

52

633 (49%)

580 (45%)

45.3

4. Saxena et al. (2010)

Prospective cohort study

63

38%

62%

55

45 (71%)

18 (29%)

NS

5. Elias et al. (2010)

Retrospective cohort study

523

43%

57%

54

370 (71%)

153 (29%)

45

6. Passot et al. (2012)

Retrospective cohort study

120

50%

50%

53,4

90 (75%)

30 (25%)

58.2

7. Ilhemelandu et al. (2013)

Retrospective cohort study

387

45.7%

55.3%

53,4

124 (32.8%)

254 (67.3%)

NS

8. Ceelen et al. (2014)

Prospective cohort study

166

50%

50%

60.2

35 (21%)

105 (63%)

18

TABLE 1 Study and patient characteristics of the eight included studies in this review. Abbreviations: ACT = Adjuvant chemotherapy Article no.

OS (months)

Median survival of ACT+ patients (months)

Median survival of ACT- patients (months)

P-value in USA

95% CI

P-value in MSA

95% CI

1.Glehen Aet al. (2004)

19.2

19.2

20.4

0.35

NR

0.01

NR

2. Shen et al. (2008)

34

15.1

22.2

0.21

ACT+: 12.1-20.3 ACT-: 18.2-37.3

>0.05

NR

3. Glehen et al. (2010)

45.3

31

45

0.008

NR

>0.05

NR

4. Saxena et al. (2010)

NR

NR

NR

>0.05

NR

>0.05

NR

5. Elias et al. (2010)

30.1

30

30

0.85

NR

>0.05

NR

6. Passot et al. (2012)

36.2

40%

15%*

0.040

NR

>0.05

NR

7. Ilhemelandu et al. (2013)

NR

NR

NR

>0.05

NR

>0.05

NR

8. Ceelen et al. (2014)

27

22

25

>0.05

ACT+: 12.9-31.1 ACT-: 19.1-30.9

>0.05

NR

TablE 2 Survival data extracted from the eight articles. * 5-year overall survival (%) AMSj Vol. 10 | February 2018


32 ARTICLE Article no.

ACT

No. of patients without morbidity

No. of patients with morbidity

P-value in UA

95% CI

P-value in MA

95% CI

4. Saxena et al. (2010)

+

29

16

0.84

NR

NR

NR

-

14

4

+

35

89

0.13

NR

0.09

OR: 0.4 0.1-1.1

-

92

162

7. Ilhemelandu et al. (2013)

Article no.

No. of patients without postoperative mortality

No. of patients with post-operative mortality

P-value in UA

95% CI

P-value in MA

95% CI

4. Saxena et al. (2010)

NR*

NR*

NR

NR

NR

NR

NR*

NR*

114

10

0.54

NR

NR

NR

234

20

7. Ilhemelandu et al. (2013)

TablE 4 Outcomes Abbreviations: UA = Univariate Analysis, MA = Multivariate Analysis * No perioperative deaths Article no.

30-day mortality after CRS and HIPEC (%)

30-day morbidity after CRS and HIPEC (%)

1.Glehen Aet al. (2004)

4.0%

22.9%

2. Shen et al. (2008)

5.5%

42%

3. Glehen et al. (2010)

4.5%

33.6%

4. Saxena et al. (2010)

0%

32%

5. Elias et al. (2010)

3%

31%

6. Passot et al. (2012)

3%

34%

7. Ilhemelandu et al. (2013)

7.7%

62%

8. Ceelen et al. (2014)

2.4%

35%

taBLE 3 Morbidity and mortality rates after CRS and HIPEC extracted from the eight articles *No perioperative deaths

an overview of the outcomes extracted from two studies analysing the specific influence of adjuvant chemotherapy on morbidity and mortality after CRS and HIPEC. We extracted data from three prospective cohort studies and five retrospective cohort studies,

with a median follow-up ranging from 18 to 86 months. The total number of participants in each study varied from 63 to 1290. The overall survival of all patients included in the studies ranged from 19.2 to 30 months (Table 1) and the median age of patients included in the selected studies ranged from 51 to 61 years. Treatment with adjuvant therapy for their primary tumour varied between 21% and 75% of the patients. The exact number of patients treated with adjuvant chemotherapy per study is shown in Table 1. One retrospective cohort study by Glehen et al. (2004) investigated the efficiency of the HIPEC procedure in 506 patients with PC and assessed possible prognostic indicators to gain more evidence regarding patient selection. Patients who were treated with adjuvant chemotherapy after their primary colon carcinoma had a median survival of 19.2 months compared to 20.4 months for patients who did not receive adjuvant chemotherapy (P=0.01) in multivariate analysis. The overall survival in this patient group was 19.2 months. The use of adjuvant chemotherapy was associated with decreased survival in HIPEC patients. Multivariate analysis also reported the completeness of cytoreduction (P<.0001), treatment with a second HIPEC procedure (P<0.001), extent of carcinomatosis (P<0.001), lymph node involve-

February 2018 | AMSj Vol. 10


33 ARTICLE

ment (P<0.002), tumour differentiation (P<0.003) and synchronous resection of liver metastasis (P=0.004) to be independent prognostic indicators of survival after CRS and HIPEC.17 Shen et al. (2008) included 121 patients with metastases to the peritoneum or liver originating from CRC. This prospective cohort study aimed to better understand the characteristics, clinical outcomes and overall survival of these patients. For our review, we only extracted data from the patients with metastases to the peritoneum. From the 55 patients with PC, 72.2% (n=40) were treated with adjuvant chemotherapy. Patients treated with adjuvant chemotherapy had a median survival of 15.1 months compared to 22.2 months in patients who did not receive adjuvant chemotherapy. However, this minor difference was not statistically significant in univariate survival analysis (P<0.21).22 In French institutions a retrospective, multi-center cohort study was performed by Glehen et al. (2010) to evaluate the efficacy and tolerance of CRS and HIPEC in patients with PC originating from non-gynecologic carcinomatosis. 1290 patients from 25 institutions were included that underwent CRS and HIPEC, of which 633 patients were treated with adjuvant chemotherapy for their primary tumour. In most cases (n=523) the PC originated from CRC. In univariate analysis treatment with adjuvant chemotherapy was associated with declined survival after CRS and HIPEC. Patients treated with adjuvant chemotherapy had a median survival of 31 months compared to 45 months in patients without such treatment (P=0.008).19 A prospective study by Saxena et al. (2010) included 63 patients and investigated the clinical and treatment-related risk factors for perioperative morbidity and mortality rates in patients with PM of CRC after CRS and HIPEC. This was the only study showing an unequal distribution of baseline characteristics, namely an unequal ratio of females and males (62% vs 38% respectively). From all patients included, 38% (n=45) received adjuvant chemotherapy for their primary colon carcinoma.

The study reported no perioperative deaths. From the 45 patients treated with adjuvant chemotherapy, 16 patients experienced severe complications compared to 4 patients in the group who did not receive adjuvant chemotherapy. However, neither univariate nor multivariate analysis showed a significant association between adjuvant chemotherapy and morbidity (P>0.05).24 A retrospective, multi-center cohort study by Elias et al. (2010) included 523 patients from 23 French centers to assess the early and longterm median survival of patients with PC from CRC after CRS and HIPEC. The majority of all included patients (n=370, 71%) received adjuvant chemotherapy for their primary colon carcinoma. This treatment with adjuvant chemotherapy was not associated with median survival both groups had a median survival of 30 months (P=0.85).21 A single-center retrospective study (n=120) by Passot et al. (2012) analysed the effect of adjuvant chemotherapy on survival in patients with PC from CRC before treatment with CRS and HIPEC. Ninety patients were treated with adjuvant chemotherapy. The use of this treatment after the primary carcinoma was correlated with improved survival in univariate analysis. The 5-year overall survival was 40% in patients who received adjuvant chemotherapy compared to 15% in patients with PC without this treatment (P=0.040). Other significant prognostic indicators on univariate analysis were age more than 50 years (P=0.025), absence of lymph node involvement (P=0.045) and complete cytoreduction (P<0.001). Multivariate analysis reported complete cytoreduction and the use of adjuvant chemotherapy after CRS and HIPEC as prognostic factors (P<0.001 and P=0.049, respectively).18 Ilhemelandu et al. (2013) investigated the prognostic value of their health-related quality of life program for predicting postoperative morbidity and mortality after CRS and HIPEC. The study retrospectively included 387 patients, 32.8% (n=124) of them received adjuvant chemotherapy following resection of their primary colon carcinoma. Univariate and multivariate analysis showed no association between treatment with adjuvant chemotherapy and morbidity or mor-

AMSj Vol. 10 | February 2018


34 ARTICLE

tality rates after CRS and HIPEC. The specific results extracted from this study are reported in Table 4.23 All other included studies reported no effect of adjuvant chemotherapy on survival after CRS and HIPEC. One prospective cohort study by Ceelen et al. (2014) investigated the effect of neoadjuvant chemotherapy with or without a biological on the survival of HIPEC patients (n=166). For our review, we excluded survival data from patients treated with a biological. The administration of adjuvant chemotherapy for the primary tumour in the group of patients that was not treated with a biological had no significant effect on survival in univariate and multivariate analysis. Patients who were treated with adjuvant chemotherapy for their primary carcinoma had a median survival of 22 months compared to 25 months in patients who were not treated with adjuvant chemotherapy (95% CI 12.9-31.1, P>0.05).20

DISCUSSION

The last decades, new treatment options and therapeutic regimes were developed to improve the prognosis of patients suffering from PM of CRC. CRS and HIPEC is a combination of approaches to remove macroscopic and microscopic tumor deposits, respectively. With this combined treatment, the median survival of these patients increased significantly from 22 months in patients who underwent CRS and HIPEC compared to 12 months in patients who received standard treatment with chemotherapy.7,25,26 Despite the potential effectiveness of the HIPEC procedure, the combined treatment is associated with relatively high morbidity and mortality rates, 15-18% and 5% respectively.7,9,12,13 Therefore, identification of prognostic factors in patients with PC is important to select those who will benefit from this treatment. This review aimed to compare the prognosis of patients with PM of CRC treated with adjuvant chemotherapy for their primary CRC with the prognosis of patients not treated with adjuvant chemotherapy. For this purpose, an extensive literature search in MEDLINE was performed. Extraction of relevant survival data from eight selected studies showed some evidence that

adjuvant chemotherapy for the primary tumour is correlated to survival of patients after CRS and HIPEC, but current evidence is not sufficient enough to guide this selection. Two studies reported a decline in median survival rates after adjuvant therapy17,19, one study showed an increase in median survival rates 18 and the remaining six articles reported no effect of adjuvant chemotherapy on the survival of HIPEC patients.20-24 The decline in median survival rates in HIPEC patients treated with adjuvant chemotherapy seen in two studies (P=0.01 and P=0.008, respectively) might be partly explained by a delay to the HIPEC procedure that might be the result of the adjuvant treatment.17,19 This could be an explanation for the shorter median survival of these patients because it jeopardizes the chance of achieving complete cytoreduction. Which is one of the most powerful prognostic indicators of survival after CRS and HIPEC. In addition, patients who received adjuvant chemotherapy for their primary colon carcinoma probably had a greater disease burden compared to those who did not receive this treatment. A third reason for the diminished survival associated with adjuvant chemotherapy might be that patients not able to respond to chemotherapy are probably also not able to respond on CRS and HIPEC. These patients, theoretically, might have more aggressive tumours than patients that do respond to chemotherapy.17 On the other hand, three included studies (Passot et al. (2012), Glehen et al. (2004) and Elias et al. (2010)) reported that higher 5-years survival rates were seen in HIPEC patients treated with adjuvant chemotherapy for their primary colon carcinoma (P=0.040) compared to HIPEC patients not treated with adjuvant chemotherapy. One reason for this observation might be that adjuvant chemotherapy probably prevents the development of metastases to other organs. Especially patients with PC from CRC are at high risk to develop metastases to the lung, liver and bones. However, the results from these studies were based on a group of patients with favorable tumour characteristics with a good chemotherapeutic response. Patients who developed metastases to the liver, lung or bone were excluded from treatment with CRS and

February 2018 | AMSj Vol. 10


35 ARTICLE

HIPEC.18 Interestingly, the results of the study of Passot et al. (2012) and those of Klaver et al. (2010) reported no significant correlation between survival and development of PC regardless of treatment with adjuvant chemotherapy. This suggests that development of PC regardless of adjuvant chemotherapy should not be an absolute contraindication to undergo CRS and HIPEC with curative intent.27 Limitations

There are several limitations in this review. First, we were only able to extract data from mostly retrospective studies and only three prospective cohort studies. Currently, there are limited phase II or phase III clinical trials available investigating the prognostic factors of patients with PC from CRC. The second limitation of this study is the we were only able to include articles written in English, we could have missed relevant articles based on language criteria. Further research is required to investigate the effect of adjuvant chemotherapy in patients who have a good response compared to those with a poor response to chemotherapy. The majority of the results are based on retrospective cohort studies, which makes the value of these hypothesis debatable. Clinical trials including larger numbers of patients with different tumour characteristics are needed in order to carefully select those with the best survival outcome after CRS and HIPEC.

CONCLUSIONS

The combination of CRS and HIPEC is an effective approach to extent the overall survival of patients with peritoneal metastases of CRC. However, the risks should not exceed the potential benefits of this combined treatment, which makes an accurate selection based on prognostic factors of patients crucial. This review was conducted to evaluate the prognosis of patients who received adjuvant chemotherapy for their primary colon carcinoma. Regarding this subject, the results of the included studies are contradictory and data are largely derived from retrospective databases. Therefore, a large prospective study focusing on survival of HIPEC patients that receive adjuvant therapy for their primary tumour should be

performed, accounting for all possible prognostic indicators. Ideally, to confirm or reject the possible role of adjuvant chemotherapy in selection of HIPEC patients, this should be followed by a randomized controlled trial. We have to base our conclusion on the existing evidence, with the majority of studies showing no association between adjuvant chemotherapy and survival rates. Therefore, there is no decisive argument to exclude patients that developed PC regardless of adjuvant chemotherapy from HIPEC treatment.

REFERENCES

1.

Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74–108. 2. Elferink MAG, de Jong KP, Klaase JM, Siemerink EJ, de Wilt JHW. Metachronous metastases from colorectal cancer: a population-based study in North-East Netherlands. Int J Colorectal Dis. 2015;30:205–12. doi: 10.1007/s00384-014-2085-6. 3. http://www.asco.org/practice-guidelines/quality-guidelines/guidelines/gastrointestinal-cancer 4. Koppe MJ, Boerman OC, Oyen WJG, Bleichrodt RP. Peritoneal carcinomatosis of colorectal origin. Ann Surg. 2006;243:212–22. doi:10.1097/01. sla.0000197702.46394.16. 5. Siegel R, Naishadham D, Jemal A. Cancer statistics, (2013). CA Cancer J Clin. 2013;63:11–30. 6. Brodsky JT, Cohen a M. Peritoneal seeding following potentially curative resection of colonic carcinoma: implications for adjuvant therapy. Dis Colon Rectum. (1991);34:723–727. 7. Verwaal VJ, van Ruth S, de Bree E, van Sloothen GW, van Tinteren H, Boot H, Zoetmulder FA (2003) Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 21(20):3737–3743. 8. Verwaal VJ, Kusamura S, Baratti D, Deraco M (2008) The eligibility for local-regional treatment of peritoneal surface malignancy. J Surg Oncol 98(4):220–223. 9. Cao Y, Tan A, Gao F, Liu L, Liao C, Mo Z (2009) A meta-analysis of randomized controlled trials comparing chemotherapy plus bevacizumab with chemotherapy alone in metastatic colorectal cancer. Int J Colorectal Dis 24(6):677–685. 10. Elias D, Gilly F, Boutitie F, Quenet F, Bereder JM, Mansvelt B, Lorimier G, Dube P, Glehen O (2010) Peritoneal colorectal carcinomatosis treated with surgery and perioperative intraperitoneal chemotherapy: retrospective analysis of 523patients from a multicentric French study. J Clin Oncol 28(1):63–68. 11. Kuijpers AM, Mirck B, Aalbers AG, Nienhuijs SW, de Hingh IH, Wiezer MJ, van Ramshorst B, van Ginkel RJ, Havenga K, Bremers AJ, de Wilt JH, te Velde EA et al (2013) Cytoreduction and HIPEC in the Netherlands: nationwide long-term outcome following the Dutch protocol. Ann Surg Oncol 20(13):4224–4230.

AMSj Vol. 10 | February 2018


36 ARTICLE 12. Maggiori L, Elias D. Curative treatment of colorectal peritoneal carcinomatosis: current status and future trends. Eur J Surg Oncol. 2010;36:599–603. doi:10.1016/j.ejso.2010.05.007. 13. Verwaal VJ, Zoetmulder FAN. Follow-up of patients treated by cytoreduction and chemotherapy for peritoneal carcinomatosis of colorectal origin. Eur J Surg Oncol. 2004;30:280–5. doi:10.1016/j.ejso.2003.12.003. 14. www.oncoline.nl 15. Sloothaak DA, Mirck B, Punt CJ, et al. Intraperitoneal chemotherapy as adjuvant treatment to prevent peritoneal carcinomatosis of colorectal cancer origin: a systematic review. Br J Cancer. 2014;111:1112–21. 16. Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group (2009). Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med 6(6):e1000097. doi:10.1371/journal. 17. Glehen O, Kwiatkowski F, Sugarbaker PH, et al. Cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multi-institutional study. J. Clin Oncol 2004; 22:3284-3292. 18. Passot G, Vaudoyer D, Cotte E, et al. Progression following neoadjuvant systemic chemotherapy may not be a contraindication to a curative approach for colorectal carcinomatosis. Ann Surg 2012; 256:125-129. 19. Glehen O, Gilly FN, Boutitie F, Bereder JM, Quenet F, Sideris L, Mansvelt B, Lorimier G, Msika S, Elias D. Toward curative treatment of peritoneal carcinomatosis from nonovarian origin by cytoreductive surgery combined with perioperative intraperitoneal chemotherapy: a multi-institutional study of 1,290 patients. Cancer. 2010;116:5608-5618. 20. Ceelen W, Van Nieuwenhove Y, Putte DV, Pattyn P. Neoadjuvant chemotherapy with bevacuzimab may improve outcome after cytoreduction and hyperthermic intraperitoneal chemoperfusion (HIPEC) for colorectal carcinomatosis. Ann Surg Oncol. 2014;21-3023-8. 21. Elias D, Gilly F, BOutitie F, et al. Peritoneal colorectal carcinomatosis treated with surgery and perioperative intraperitoneal chemotherapy: retrospective analysis of 523 patients from a multicentric French study. J Clin Oncol 2010; 28:63-68. 22. Shen P, Thai K, Stewart JH, et al. Peritoneal surface disease from colorectal cancer: comparison with the hepatic metastases surgical paradigm in optimally resected patients. Ann Surg Oncol 2008; 15:3422-3432 23. Ilhemelandu CU, McQuellon R, Shen P, et al. Predicting postoperative morbidity following cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CS + HIPEC) with Preoperative FACT-C (functional assessment of cancer therapy) and patient-rated performance status. Ann Surg Oncol 2013; 20:3519-3526. 24. Saxena A, Yan TD, Morris DL. A critical evaluation of risk factors for complications after cytoreductive surgery and perioperative intraperitoneal chemotherapy for colorectal peritoneal carcinomatosis. World J Surg 2010; 34:70-78. 25. Shen P, Hawksworth J, Lovato J, et al. Cytoreductive surgery and intraperitoneal hyperthermic chemotherapy with mitomycin C for peritoneal carcinomatosis from nonappendiceal colorectal carcinoma. Ann Surg Oncol 2004; 11(2):178-86.

26. Verwaal VJ, Bruin S, Boot H, et al. 8-year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol 2008; 15(9):2426-32. 27. Klaver YL, de Hingh IH, Boot H, et al. Results of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy after early failure of adjuvant systemic chemotherapy. J Surg Oncol. 2010;103:431-434.

February 2018 | AMSj Vol. 10


37 EXPERT OPINION

Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease P.J. Hengeveld and G. de Waard

Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease, MS Sabatine et al., FOURIER Steering Committee and Investigators, New England Journal of Medicine, 2017, 17 March

Abstract

B a c kg r o u n d Evolocumab is a monoclonal antibody that inhibits PCSK9 and lowers LDL cholesterol levels by approximately 60%. Whether it prevents cardiovascular events is uncertain. M e t h o d s We conducted a randomized, double-blind, placebo-controlled trial involving 27,564 patients with atherosclerotic cardiovascular disease and LDL cholesterol levels of 70 mg per deciliter (1.8 mmol per liter) or higher who were receiving statin therapy. Patients were randomly assigned to receive evolocumab (either 140 mg every 2 weeks or 420 mg monthly) or matching placebo as subcutaneous injections. The primary efficacy end point was the composite of cardiovascular death, myocardial infarction, stroke, hospitalization for unstable angina, or coronary revascularization. The key secondary efficacy end point was the composite of cardiovascular death, myocardial infarction, or stroke. The median duration of follow-up was 2.2 years. RESULTS At 48 weeks, the least-squares mean percentage reduction in LDL cholesterol levels with evolocumab, as compared with placebo, was 59%, from a median baseline value of 92 mg per deciliter (2.4 mmol per liter) to 30 mg per deciliter (0.78 mmol per liter) (P<0.001). Relative to placebo, evolocumab treatment significantly reduced the risk of the primary end point (1344 patients [9.8%] vs. 1563 patients [11.3%]; hazard ratio, 0.85; 95% confidence interval [CI], 0.79 to 0.92; P<0.001) and the key secondary end point (816 [5.9%] vs. 1013 [7.4%]; hazard ratio, 0.80; 95% CI, 0.73 to 0.88; P<0.001). The results were consistent across key subgroups, including the subgroup of patients in the lowest quartile for baseline LDL cholesterol levels (median, 74 mg per deciliter [1.9 mmol per liter]). There was no significant difference between the study groups with regard to adverse events (including new-onset diabetes and neurocognitive events), with the exception of injection-site reactions, which were more common with evolocumab (2.1% vs. 1.6%). c o n c l u s i o n In our trial, inhibition of PCSK9 with evolocumab on a background of statin therapy lowered LDL cholesterol levels to a median of 30 mg per deciliter (0.78 mmol per liter) and reduced the risk of cardiovascular events. These findings show that patients with atherosclerotic cardiovascular disease benefit from lowering of LDL cholesterol levels below current targets.

Background

Low density lipoprotein (LDL) cholesterol is an important modifiable risk factor for cardiovascular disease. The recently developed monoclonal antibody evolocumab, targeting proprotein convertase subtilisin-kexin type 9 (PCSK9), has superior lipid-lowering effects to conventional statin therapy, additionally decreasing LDL cholesterol levels by approximately 60%. The trial we will discuss in this article is the first to assess the potential of evolocumab as secondary prevention for cardiovascular events in a large patient cohort.

Review

This trial is the first of its kind evaluating the efficacy of evolocumab in a large patient cohort. Key strengths include its size (analysing 59.865 cumulative patient-years), it’s randomized, double-blind, placebo-controlled nature and the inclusion of necessity of intervention (such as hospitalization or coronary revascularisation) as additional end points. An additional strength is its intention-to-treat design, employing routinely prescribed secondary preventative pharmacotherapy alongside evolocumab (not mentioned in the abstract).

AMSj Vol. 10 | February 2018


38 clinical image

An important shortcoming of this trial is the relatively short median duration of follow-up of 2.2 years. As the risk of cardiovascular events in this patient population is elevated into the indefinite future, the relatively small effect size reported in this study, an absolute risk-reduction of merely 1.5%, might increase over an extended duration of treatment.

Conclusion

Overall, we assess this study as high-quality scientific work, providing interesting data and cause for discussion to the field of secondary prevention for cardiovascular events. Research assessing the long term efficacy of evolocumab treatment is warranted.

Answers A. Zwagemaker, M.J.S. Oosterveld A. Genitourinary tuberculosis Genitourinary tuberculosis is commonly seen in developing countries and may lead to urinary tract infections and nephritides. The diagnosis is established by detection of tubercle bacilli in urine and heterogeneous imaging findings. These include signs of chronic pyelonephritis, papillary necrosis and scarring of renal tissue.1 A plain abdominal radiograph may demonstrate localized calcifications, but usually not generalized involvement as seen on the X-ray of our patient. B. Nephrocalcinosis Our patient suffered from nephrocalcinosis. The most striking finding on his abdominal X-ray is nephrocalcinosis, which is characterized by generalized calcium deposition in the kidney. Calcification occurs when urinary excretion of calcium phosphate or oxalate is increased. Causes include primary hyperparathyroidism, sarcoidosis and a range of inherited conditions.2 In our patient a homozygous mutation in the AGXT-gen was detected, confirming a diagnosis of primary hyperoxaluria type 1. This is a rare autosomal recessive defect of hepatic oxalate metabolism leading to nephrocalcinosis and recurrent kidney stones.3 As a result of advanced renal disease, our patient suffered from renal osteodystrophy and (uremic) polyneuropathy. His clinical condition was deemed too poor for liver transplantation and the patient passed away at the age of 18 years. C. Nephrolithiasis Kidney stones are related to, but not the same as, nephrocalcinosis. Whereas nephrocalcinosis reflects calcification of renal tissue, nephrolithiasis is stone formation within the renal collecting system. Therefore, abdominal X-ray would depict stones within the collecting system and no generalized calcification. D. Urolithiasis In contrast to nephrolithiasis, urolithiasis refers to stones within the urinary tract. Typically, stones are formed in the kidney and lead to symptoms when they pass the ureter. Clinical manifestations include renal colic pain and gross hematuria. Both nephrolithiasis and urolithiasis are much more common compared to nephrocalcinosis.

References

1. 2. 3.

Pais VM, Dionne-Odom J. UpToDate: Renal disease in tuberculosis. Last updated May 02, 2017. Kobrin SM. UpToDate: Nephrocalcinosis. Last updated April 17, 2017. Strauss SB, Waltuch T, Bivin W et al. Primary hyperoxaluria: spectrum of clinical and imaging findings. Pediatr Radiol, 2017; 47(1): 96-103.

February 2018 | AMSj Vol. 10


39 ARTICLE

Thinking about tomorrow: A literature review on the long-term impact of adolescent cannabis use on cognition and intelligence T.E.K. van Os, medical student, VU University Medical Center, Amsterdam T.J. de Vries, Department of Anatomy and Neuroscience, VU University Medical Center, Amsterdam

Abstract

I n t r o d u c t i o n Cannabis is the most used illicit drug among adolescents. Previous studies showed an association between cannabis use and lower white matter volume and decreased cognitive performance. The aim of this literature review is to give a detailed overview of studies assessing the long term effects of cannabis use by adolescents (13-24 year old) in cognitive functioning and intelligence. M e t h o d s A literature search was performed and 16 relevant studies were found. These studies measured the long lasting effect on five of the six neurocognitive domains (complex attention, executive function, learning and memory, language, perceptual-motor function) and overall intelligence (IQ). Exclusion criteria were co-morbid psychiatric problems and pre-adolescent cannabis exposure. RESULTS Overall findings in the studies showed that persistent adolescent cannabis users performed worse than non-cannabis users on executive function, memory, learning, psychomotor function and IQ, but not on attention and perceptual-motor function. Earlier onset is associated with worse performance of attention, executive function, memory and IQ. Furthermore, frequency of cannabis use was associated with a decline in attention, executive function, memory, psychomotor speed and IQ, however not in perceptual-motor function. Abstinence did not improve attention and short-term memory, but did improve learning and executive functioning. Additionally, alcohol co-abuse was no significant predictor for a deficit in any domain. c o n c l u s i o n Earlier onset and higher frequency are associated with deficits in cognitive functioning. However, studies assessing persistent long term effect of cannabis after adolescence are scarce. Thus, more longitudinal studies are important. Concluding, this review notifies a greater public awareness is needed to prevent adolescents from frequent and persistent cannabis use, and to delay the onset of cannabis use.

INTRODUCTION

Cannabis is the most used illicit drug worldwide.1, 2 In the United States, the annual prevalence of cannabis use by adolescents is 23.7%. The daily use rates in 8th (13-14 years old), 10th (15-16 years old), and 12th (17-18 years old) grades were 1.1%, 3.0%, and 6.0%, respectively.3 In the Netherlands, monthly prevalence of cannabis use reached a peak in the young adults (20-24 years old) group (10.3%). And although drug use prevalence is highest among people in their twenties, in comparison to hard drugs (cocaine, ecstasy or amphetamine), cannabis use is relatively high in the adolescent group.4 While cannabis use is common among adolescents, cannabis use may have long lasting effects on neurocognitive functioning, specifically in adolescents.5 This might be due to the fact that adolescence is marked by brain development. From the beginning of puberty until 24 years old,

the brain rewires itself. This is accomplished by myelination, which increases the speed of impulse conduction across these neurons, and eradication of unused synapses. This process takes place especially in the prefrontal cortex of adolescents, since the prefrontal cortex is one of the last regions in which brain development is completed. The strengthening of the neurocircuitry enhances multitasking, the capability of processing complex information and problem solving. Additionally, the neuroplasticity of the adolescent’ brain provides the possibility to learn and adapt. However, due to this sensitive period in brain development, drug abuse could have a negative effect to the brain maturation.6 There is epidemiological evidence that cannabis use is a risk factor for schizophrenia and psychosis, and that in individuals with schizophrenia, using cannabis results in a worsening of symptoms.7, 8 These mental disorders are

AMSj Vol. 10 | February 2018


40 ARTICLE

associated with a smaller white matter in the brain of cannabis users.5 Structural differences were found in cortical areas like the orbitofrontal region, hippocampus, amygdala and inferior posterior lobules (lobules VIII-X), which are responsible for the cognitive functions decision making, memories, emotion processing and executive functioning, respectively. Considering the sensitive period in brain development and the effect of cannabis on white matter volume in the aforementioned brain structures, a negative effect of cannabis use on cognitive functioning is expected. This hypothesis is supported by a recent systematic review, showing evidence of overall IQ, attention and memory deficits in adolescent (chronic) cannabis users.5 These deficits remit after three or more months of cessation.9 Furthermore, heavy cannabis use is more associated with long lasting effects on cognitive functioning than an earlier onset.5 This systematic review, however, did not assess the effect of cannabis use on intelligence. Moreover, early onset was defined as age of onset before 18 years. The aim of this literature review is to give a more detailed overview of studies assessing the long term effect of cannabis use by adolescents (13-24 year old), specifically on cognitive functioning and intelligence (IQ). Although intelligence and cognitive functioning are not the same, there is an overlap between these two and they are intertwined and related. Moreover, adolescence is most commonly defined as a stage including the ages of 10-18, but can also include ages of 9-26.10 In this review, adolescents were defined as individuals with an age between 13 and 24 years old, since the brain maturates up until the age of 25. This review will focus on frequency, age at onset and the duration of abstinence of cannabis use. Furthermore, co-abuse of alcohol will be taken into consideration. Research regarding harmful long term effects on cannabis use is important, considering evidence of association with white matter volume decrease, vulnerability of the brain and evidence of neurocognitive impairment among adolescent cannabis users. Considering the popularity of cannabis among adolescents and the legal age of buying cannabis (e.g. in the Netherlands it is 18 years), the primary aim of this review is to give a detailed overview of the long-term effects of cannabis use.

This may help to guide public-health discussions and further research in this area.

METHODS

A literature search was performed using PubMed and Web of Science. The use of two databases contributed to a more widespread and diverse pool of articles. In both PubMed as Web of Science, animal studies were excluded and only human studies were included. Literature and systematic reviews were used to find additional articles. Search strategy

The MeSH- and free terms ‘adolescent’ and ‘young adults’ were included. To find articles concerning cannabis, MeSH-terms ‘cannabis’, ‘marijuana abuse’ and ‘marijuana smoking’ were included. Moreover, free terms ‘cannabis’ and ‘marijuana’ were included as well. The use of these free terms also provided studies assessing alcohol co-abuse among adolescents. This review focusses on long-term effect on cognitive function and intelligence. To search for articles concerning cognitive function, MeSH-term ‘cognition’ and ‘cognitive science’ were included, as well as the free term ‘cognition’ and ‘cognitive’. Through inclusion of MeSH-terms ‘longitudinal studies’, ‘prospective studies’ and free terms ‘long term’, ‘longterm’ and ‘long-term’, studies concerning the long-term effect criteria will be acquired. Thereby, studies assessing intelligence (IQ) will be included. Study selection

Although medicinal cannabis could have an effect on brain maturation, the MeSH-term ‘medicinal marijuana’ and ‘treatment outcome’ were used as exclusion criteria. Medicinal cannabis is not fully representative for common used cannabis. This is due to a different ratio of CBD and THC in medicinal cannabis in comparison to street cannabis. Moreover, to focus on adolescents, studies assessing pre-adolescent and prenatal cannabis exposure are excluded. Likewise, to have a representative population, studies concerning individuals with co-morbid psychiatric problems were eliminated, knowing that there is a correlation between smoking cannabis and psychiatric problems. Furthermore, since the focus is not on

February 2018 | AMSj Vol. 10


41 ARTICLE

acute effects of cannabis use, studies concerning short-term effects are excluded. An additional search was performed in PubMed and Web of Science using a combination of free terms, to make sure all relevant articles were found. This combination consisted of the free terms ‘long term effects cannabis marijuana adolescents’. The search strategy provided 95 hits. Each article title and some abstracts were reviewed. The additional search of free terms gave 62 hits in Web of Science and almost 900 hits in PubMed. The titles and some abstracts of the 62 hits in Web of Science and the first 100 hits in PubMed, sort by ‘Best Match’, were reviewed for in- and exclusion criteria. Combining the two strategies and the acquired articles using systematic and literature reviews, provided 15 relevant studies. One additional article, which was submitted online after the first search, was added last minute, making a total of 16 relevant studies (see supplementary FILE 1 online). Additionally, systematic and literature reviews were found and analysed. The studies used in these reviews matched the articles found using the search strategy explained above.

RESULTS

The articles were categorized by six neurocognitive domains, which DSM-V used to define cognitive functions.11 These domains are (1) complex attention, (2) executive function, (3) learning and memory, (4) language, (5) perceptual-motor function and (6) social cognition, each of these domains further divided in multiple subdomains. Since there were no studies performed measuring social cognition, this domain will not be featured in this review. However, as mentioned in the Methods-section, overall intelligence (IQ) will be discussed. Complex attention

Seven studies examined differences in complex attention. The majority of these studies provided evidence for attention deficits, although some were subtle. Early-onset (between 12 years and 16 years old) is a predictor for deficit in reaction times, specific to visual scanning (using cannabis 3-4 times a week). Late-onset cannabis (above age 16) users did not differ from controls.12 Stud-

ies done among adolescent cannabis users showed that cannabis use had a subtle negative effect on two measurements: Rapid Visual Information Processing test (smoking once a week) and Immediate Memory Task (smoking average 4 times a week).13, 14 A 3-year follow-up study, assessing cannabis use and co-abuse of alcohol in adolescents, found that 1 of 8 tests were significant for attention impairment due to cannabis and alcohol use.15 However, increased lifetime cannabis use among adolescents was associated with poorer attention performance as well.16 Even with abstinence of 3 weeks, persistent adolescent cannabis users showed decreased attention accuracy.17 A study done following individuals from birth to age 38, asked the participants to let people “who knew them well” fill in questionnaires and checklists. Informants provided evidence that participants with more persistent cannabis dependency showed significantly more attention problems.18 Executive function

Nine studies assessed the effect of cannabis use on executive function. Only one of the studies did not find any effect in this domain after a 3-year follow-up study performed among adolescent participants with a co-abuse of alcohol.15 One study (participants aged 14-18, average cannabis use five times a week) provided evidence that decision making was not impaired, but there is evidence of an increase in impulsivity.13 On the other hand, an association between greater impairment in executive functions was associated with a higher amount of cannabis use and more persistent cannabis dependency among adolescents.14, 18, 19 Furthermore, early-onset (age below 16) chronic (heavy) cannabis users performed poorly on executive functions compared to controls and late-onset cannabis users.19, 20 However, an improvement after abstinence of 3 weeks, for study purposes, was seen.17 Learning and memory

Ten studies examined the effects of cannabis use on learning and memory. Compared to adolescent non-cannabis users, frequent (4 times per week) adolescent cannabis users performed worse on short-term memory.13 Poor performance on shortterm memory was associated with a younger onset of cannabis use,21 specifically an age of

AMSj Vol. 10 | February 2018


42 ARTICLE

onset under the age of 15 years and 17 years. 22 Besides age of onset, also duration, frequency and lifetime cannabis use was associated with a greater impairment,21 even with 28 days of abstinence.16 However, heavy users, using cannabis daily, improved their performance after 3 months of voluntary cannabis cessation 23 and weekly cannabis users improved their performance after 1 year of stopping cannabis use.24 Furthermore, persistent cannabis users aged 15-19 performed worse than controls in learning, but improved after abstinence of 2 and 3 weeks.17 On the contrary, one study found an association between higher lifetime alcohol co-abuse episodes and better memory performance.16 However, this association was not seen in another study assessing alcohol co-abuse.17 Moreover, a study done following individuals from birth to age 38, asked the participants to let people “who knew them well� fill in questionnaires and checklists. Informants provided evidence that participants with more persistent cannabis dependency showed significantly more memory problems.18 20,

Language

Two studies examined verbal intelligence in adolescent-onset users. One study, following participants men from ages 13 to 20 years, found a relation between higher verbal IQ and an earlier age of onset (before the age of 15) and an increase in cannabis use frequency.20 On the contrary, in another study, reviewing subjects at age 30-55 years, early-onset cannabis users (before the age of 17) had a lower verbal IQ-score in comparison to controls.22 Perceptual-motor function

Two studies assessed the effect of adolescent cannabis use on perceptual-motor function. Both studies did not see a significant correlation between cannabis use and performance on visuospatial function.15, 16 However, one of these two studies did find an association between psychomotor speed and increased lifetime cannabis use: there was an correlation between a better psychomotor speed and an increased lifetime alcohol consumption.16 Intelligence

Five studies examined differences in IQ-scores.

A greater IQ decline was found in early-onset cannabis users (age of onset before 18 years) in comparison to late-onset cannabis users.18 Also, subjects with more persistent cannabis abuse showed a greater IQ decline.18 Furthermore, IQscores in current heavy cannabis users (smoking at least 5 joints a week), at the age of 17-21, were significantly different compared to non-users.23, 25 However, in these two studies, no significant difference in light current users (less than 5 joints a week), former users and non-cannabis users was found. While heavy cannabis use is associated with a IQ decline, former heavy cannabis users, who were voluntary abstinent for at least three months and smoked at least 5 joints a week prior to the abstinence, did not show a significant difference in IQ performance compared to non-users.25 Although these three studies saw a difference in IQ associated with cannabis use, two other studies did not see this correlation. One found no significant difference in IQ between early-onset, late-onset and non-cannabis users.26 Another study, the largest longitudinal study on cannabis use and IQ performance, did find a greater decline in cannabis users in comparison to non-users.27 However, this effect is debatable due to a lack of difference between siblings, the presence of baseline differences before cannabis onset and the absence of a dose–response relationship. Therefore, this decline could be explained by confounding factors.27

DISCUSSION

Overall, the reviewed studies concluded that executive function, short-term memory, learning and psychomotor function were significantly worse in persistent adolescent cannabis users in comparison to non-cannabis users. In contrast to complex attention and perceptual function, where no or subtle significant differences between users and non-users were found. Although a greater decline in IQ-score was found, this effect was debatable. An earlier onset of cannabis use, ranging between 14 and 17, was a predictor for a worse performance of complex attention, executive function, short-term memory and IQ in some studies. Furthermore, frequency of cannabis use was associated with decreased performance in areas of complex attention, executive function, short-term memory, psychomotor speed and IQ, however not

February 2018 | AMSj Vol. 10


43 ARTICLE

in perceptual function. Abstinence of three weeks did not improve performance in complex attention and short-term memory, but did improve learning and executive functioning. Cessation of cannabis for more than 3 months showed a normalisation of short-term memory performance in former heavy users. Additionally, co-abuse of alcohol was assessed in some studies. No significant worsening of performance due to co-abuse was found in these studies, comparing cannabis users to cannabis and alcohol users. Unexpectedly, a higher frequency of alcohol use predicted a better performance on short-term memory and psychomotor speed. At last, an unpredicted relation was seen in verbal IQ. Early-onset and increase in cannabis frequency did show a higher score on verbal IQ. These conclusions are globally consistent with previous reviews, although these reviews analysed studies using adult participants as well. There are two main differences seen between studies reviewed in this review and those assessed in previous reviews.5, 9 First, differences found in attention between cannabis users and non-cannabis users were subtle in this review, while these differences where more evident in previous reviews. Second, in previous reviews, heavy cannabis use among adolescents was more associated with an effect than early-onset, on diverse domains. In this review, early-onset and heavy cannabis use were roughly equal predictors for neurocognitive impairment. This could be explained by the fact that the average age of participants in the majority of the studies analysed in this review were between 15 and 20 years. These teenagers have a lower lifetime cannabis use and use cannabis less frequent compared to people in their twenties.4 The average age of the population is one major limitation in these studies. Because the average age of participant were between 15 and 20 years, a statement regarding long-term effects is hard to make. Since brain development continues until mid-twenties, long-term effect should be assessed after that age as well. However, studies assessing cannabis users older than 25 years are scarce. Therefore, animal studies could have been reviewed. By using animal studies, the long-term effect of cannabis could be measured more precisely, since confounding

factors, cannabis use and abstinence can be controlled for. Besides the average age of participants in the reviewed studies, some more limitations in the studies were found. One is the lack of assessing abstinence periods. Out of sixteen studies, three studies measured cognitive functioning after cessation of 3 weeks and two studies assessed impairment of former heavy and former light cannabis users. There was also a widespread variation in cannabis use among participants. Not only monthly or weekly frequency, but lifetime cannabis use as well. Thereby, most studies categorized heavy, regular or light smokers by a different cannabis use frequency. Where one study defines a heavy smoker as using cannabis at least once a week, another study describes a light smoker as using cannabis fewer than 5 times a week. Due to this variation, a safe frequency of cannabis use is hard to define. Another limitation found in the studies was the lack of focus on nicotine use in combination with cannabis. This is important because recent studies show an association between adolescent nicotine intake and impaired cognitive function.28-30 The lack of focus might be due to the fact that Americans use cannabis more commonly without tobacco, while in Europe cannabis users tend to add tobacco to the cannabis.31 Considering these limitations found in the reviewed studies, more (longitudinal) studies are important to analyse the long-term effect of cannabis on adolescents’ brains. By following participants from age 10 to 30 and up, and administrating a battery of tests measuring cognitive functioning and intelligence every couple years, there might be a possibility to determine which impairments or abnormalities are caused by cannabis use. Monitoring cannabis use every couple months should give a better insight into consequences of frequency and age of onset on cognitive functioning and intelligence. Moreover, potential improvement after discontinuing cannabis use, and at what age, could be analysed. By using a widespread and large population, possible confounding or effect modification, such as substance co-abuse, pre-puberty intelligence, socioeconomic status or psychiatric problems, could be analysed. Although inconsistencies in the reviewed studies remain, some evidence was found that using

AMSj Vol. 10 | February 2018


44 ARTICLE

cannabis in adolescence has a negative long-term effect on cognitive functioning and intelligence. Especially an earlier onset, using cannabis before the age of 18, and a more frequent use of cannabis are predictors for cognitive impairment and intelligence. Thereby, abstinence may improve performance in some domains, but not in all. A greater public awareness is needed to prevent adolescents from frequent and persistent cannabis use, and to delay the onset of cannabis use.

REFERENCES

1.

2. 3.

4. 5.

6. 7.

8.

9. 10. 11.

12. 13.

14.

Degenhardt L, Whiteford HA, Ferrari AJ, et al. Global burden of disease attributable to illicit drug use and dependence: findings from the Global Burden of Disease Study 2010. Lancet (London, England). 2013;382(9904):1564-74. Winstock AR. The Global Drug Survey 2015 findings. 2015. Johnston LD, O’Malley, P. M., Miech, R. A., et al., Monitoring the Future national survey results on drug use, 1975-2015: Overview, key findings on adolescent drug use. 2016. van der Pol P, & van Laar, M. Kerncijfers drugsgebruik 2014. 2015. Ganzer F, Bröning S, Kraft S, et al. Weighing the Evidence: A Systematic Review on Long-Term Neurocognitive Effects of Cannabis Use in Abstinent Adolescents and Adults. Neuropsychology Review. 2016;26(2):186222. Arain M, Haque M, Johal L, et al. Maturation of the adolescent brain. Neuropsychiatric Disease and Treatment. 2013;9:449-61. Malone DT, Hill MN, Rubino T. Adolescent cannabis use and psychosis: epidemiology and neurodevelopmental models. British Journal of Pharmacology. 2010;160(3):511-22. van Winkel R, Kuepper R. Epidemiological, neurobiological, and genetic clues to the mechanisms linking cannabis use to risk for nonaffective psychosis. Annual review of clinical psychology. 2014;10:767-91. Jacobus J, Bava S, Cohen-Zion M, et al. Functional Consequences of Marijuana Use in Adolescents. Pharmacology, biochemistry, and behavior. 2009;92(4):559-65. Curtis AC. Defining Adolescence. Journal of Adolescent and Family Health. 2015;7(2). American Psychiatric Association. Neurocognitive Disorders. Diagnostic and Statistical Manual of Mental Disorders 5th ed. Washington, DC: http://dx.doi. org/10.1176/appi.books.9780890425596.dsm17; 2013. Ehrenreich H, Rinn T, Kunert HJ, et al. Specific attentional dysfunction in adults following early start of cannabis use. Psychopharmacology. 1999;142(3):295-301. Dougherty DM, Mathias CW, Dawes MA, et al. Impulsivity, Attention, Memory, and Decision-Making among Adolescent Marijuana Users. Psychopharmacology. 2013;226(2):307-19. Harvey MA, Sellman JD, Porter RJ, et al. The relationship between non-acute adolescent cannabis use and

cognition. Drug and alcohol review. 2007;26(3):309-19. 15. Jacobus J, Squeglia LM, Infante MA, et al. Neuropsychological performance in adolescent marijuana users with co-occurring alcohol use: A three-year longitudinal study. Neuropsychology. 2015;29(6):829-43. 16. Medina KL, Hanson KL, Schweinsburg AD, et al. Neuropsychological functioning in adolescent marijuana users: subtle deficits detectable after a month of abstinence. Journal of the International Neuropsychological Society : JINS. 2007;13(5):807-20. 17. Hanson KL, Winward JL, Schweinsburg AD, et al. Longitudinal study of cognition among adolescent marijuana users over three weeks of abstinence. Addictive behaviors. 2010;35(11):970-6. 18. Meier MH, Caspi A, Ambler A, et al. Persistent cannabis users show neuropsychological decline from childhood to midlife. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(40):E2657-64. 19. Gruber SA, Sagar KA, Dahlgren MK, et al. Age of onset of marijuana use and executive function. Psychology of addictive behaviors : journal of the Society of Psychologists in Addictive Behaviors. 2012;26(3):496-506. 20. Castellanos-Ryan N, Pingault J-B, Parent S, et al. Adolescent cannabis use, change in neurocognitive function, and high-school graduation: A longitudinal study from early adolescence to young adulthood. Development and Psychopathology. 2016:1-14. 21. Solowij N, Jones KA, Rozman ME, et al. Verbal learning and memory in adolescent cannabis users, alcohol users and non-users. Psychopharmacology. 2011;216(1):13144. 22. Pope HG, Jr., Gruber AJ, Hudson JI, et al. Early-onset cannabis use and cognitive deficits: what is the nature of the association? Drug and alcohol dependence. 2003;69(3):303-10. 23. Fried PA, Watkinson B, Gray R. Neurocognitive consequences of marihuana--a comparison with predrug performance. Neurotoxicology and teratology. 2005;27(2):231-9. 24. Tait RJ, Mackinnon A, Christensen H. Cannabis use and cognitive function: 8-year trajectory in a young adult cohort. Addiction (Abingdon, England). 2011;106(12):2195-203. 25. Fried P, Watkinson B, James D, et al. Current and former marijuana use: preliminary findings of a longitudinal study of effects on IQ in young adults. CMAJ : Canadian Medical Association journal = journal de l’Association medicale canadienne. 2002;166(7):887-91. 26. Fontes MA, Bolla KI, Cunha PJ, et al. Cannabis use before age 15 and subsequent executive functioning. The British journal of psychiatry : the journal of mental science. 2011;198(6):442-7. 27. Jackson NJ, Isen JD, Khoddam R, et al. Impact of adolescent marijuana use on intelligence: Results from two longitudinal twin studies. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(5):E500-8. 28. Han G, An L, Yang B, et al. Nicotine-induced impairments of spatial cognition and long-term potentiation in adolescent male rats. Human & experimental toxicology. 2014;33(2):203-13. 29. Goriounova NA, Mansvelder HD. Short- and long-

February 2018 | AMSj Vol. 10


45 RADIOLOGY image term consequences of nicotine exposure during adolescence for prefrontal cortex neuronal network function. Cold Spring Harbor perspectives in medicine. 2012;2(12):a012120. 30. Yuan M, Cross SJ, Loughlin SE, et al. Nicotine and the adolescent brain. The Journal of physiology. 2015;593(16):3397-412. 31. Hindocha C, Freeman TP, Ferris JA, et al. No Smoke without Tobacco: A Global Overview of Cannabis and Tobacco Routes of Administration and Their Association with Intention to Quit. Frontiers in Psychiatry. 2016;7.

Answers S. Spijkers & M. Maas 1B, 2A, 3C, 4B, 5D

Reference

Adenomyosis is a form of endometriosis in which the endometrium breaks through the muscle wall of the uterus (the myometrium). As a consequence, complaints about menstrual cramps, lower abdominal pressure, and heavy elongated menstrual periods can be present in the female patient. Pelvic MRI, especially T2 weighted images, is the modality of choice to diagnose and characterise adenomyosis, with a sensitivity of 78-88% and a specificity of 67-93%. The most easily recognised feature is thickening of the junctional zone of the uterus (normal junctional zone measures up to ~5 mm). Focal thickening of the junctional zone can easily lead to a diagnosis of adenomyosis, whereas diffuse thickening should be differentiated from physiologic thickening. The presence of bright foci on T2- or T1-weighted images supports the diagnosis of adenomyosis. In general, a maximal junctional zone thickness of more than 12 mm is highly predictive of the presence of adenomyosis, while a thickness of less than 8 mm usually allows exclusion of the disease3.

Explanation of the image

The left image shows increased myometrial thickness and multiple high intensity foci inside the myometrium which are typical for adenomyosis. In the right image a normal junctional zone is seen.

References

1. 2. 3.

Roth, C. and Deshmukh, S. (2012). Fundamentals of body MRI. 1st ed. Elsevier Saunders. Gaillard, F. (2017). Adenomyosis of the uterus | Radiology Reference Article | Radiopaedia.org. [Accessed 22 Mar. 2017]. Tamai K, Togashi K, Ito T, Morisawa N, Fujiwara T, Koyama T. (2005) MR Imaging Findings of Adenomyosis: Correlation with Histopathologic Features and Diagnostic Pitfalls 1. Radiographics. Jan;25(1):21-40.

AMSj Vol. 10 | February 2018


46 ARTICLE

Colophon Amsterdam Medical Student journal is a scientific medical journal with the purpose to enable medical students to publish clinical observations, research articles and case reports. The journal was founded by students from the Academic Medical Center (AMC) and the VU University Medical Center (VUmc) in Amsterdam with the intention to provide education and development of academic skills for medical students. The entire journal is created and published by staff members and students from both faculties. ISSN 2589-1243 (print); 2589-1251 (online) CORRESPONDANCE info@amsj.nl

Cover Pharmacy medical herbs in old drawers, Krรกkow, Poland. Photo by Samuel Zeller on Unsplash. PRINT Drukwerkdeal.nl COPYRIGHT & WARRANTY Statements, opinions, and results of studies published in Amsterdam Medical Student journal are those of the authors and do not reflect the policy of the Academic Medical Center, VU University Medical Center, the Editors or Board of AMSj. The Academic Medical Center, VU University Medical Center, Editors and Board of AMSj provide no warranty as to their accuracy or reliability. This volume of Amsterdam Medical Student journal is licensed under a CC BY-NC-ND 4.0 license. For more information on your rights to share this work, please see the full lincense on bit.ly/1weyPUN

WEB www.amsj.nl Like us on facebook.com/amsjournal SUBMISSIONS If you would like to publish your research in AMSj, please see our guidelines on www.amsj.nl EDITORS IN CHIEF A. Farokhi, M.T.U. Schuijt, A.V. de Jonge, G.E. Linthorst, F. Daams BOARD K. Verdeyen, W.D. Krishnasing, A.J. Hoving, S. Veldkamp, J.W. Bruggeman, B. Lebbink CONTENT EDITOR & NATIVE EDITOR T.R. de Back, M.E. Ribbink STAFF REVIEWERS P.P. de Koning, L. Vogt, M. Kok, M. Maas, W.W.L. Li, M.W. Hollman, I.N. van Schaik, M.U. Schafroth, M.J.S. Oosterveld, V.G.M. Geukers, L.M. Hubers, J.W. Wilmink, C.G. Genc, G. de Waard, J.A.M. Bramer, W. Fuijkschot, J.S. ten Kulve, J. Cloos, S. Bossers, N. Rol, J. Tichelaar, M. Best, M.W. Bijlsma, B. de Witte, R. Holman STUDENT REVIEWERS A. van Aken, M. Arnoldussen, F. Dusseldorp, S. Spijkers, R. Betgem, A.F. Zwagemaker, M. Nazir, S. Hoeffelman, T. J. van Trier, D.E.F.W.M. van Toledo, A.R. Schuurman, R.J. Molenaar, N. Wolfhagen, J.V. Veld, A.R. Leemeijer, J. Saris, E. Leenders, M. van Baar, D.N.H. van Vemde, K. Smit, P.J. Hengeveld, M. de Jong, R.A.L. Beekman GRAPHIC DESIGN D.H. Schoenmakers

February 2018 | AMSj Vol. 10


WWW.A


AMSj.nl


Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.